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
|
Zimmerman AJ, Serrano-Rodriguez A, Sun M, Wilson SJ, Linsenbardt DN, Brigman JL, Weick JP. Knockout of AMPA receptor binding protein Neuron-specific gene 2 (NSG2) enhances associative learning and cognitive flexibility. Mol Brain 2024; 17:95. [PMID: 39695712 DOI: 10.1186/s13041-024-01158-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
The vast majority of gene mutations and/or gene knockouts result in either no observable changes, or significant deficits in molecular, cellular, or organismal function. However, in a small number of cases, mutant animal models display enhancements in specific behaviors such as learning and memory. To date, most gene deletions shown to enhance cognitive ability generally affect a limited number of pathways such as NMDA receptor- and translation-dependent plasticity, or GABA receptor- and potassium channel-mediated inhibition. While endolysosomal trafficking of AMPA receptors is a critical mediator of synaptic plasticity, mutations in genes that affect AMPAR trafficking either have no effect or are deleterious for synaptic plasticity, learning and memory. NSG2 is one of the three-member family of Neuron-specific genes (NSG1-3), which have been shown to regulate endolysosomal trafficking of a number of proteins critical for neuronal function, including AMPAR subunits (GluA1-2). Based on these findings and the largely universal expression throughout mammalian brain, we predicted that genetic knockout of NSG2 would result in significant impairments across multiple behavioral modalities including motor, affective, and learning/memory paradigms. However, in the current study we show that loss of NSG2 had highly selective effects on associative learning and memory, leaving motor and affective behaviors intact. For instance, NSG2 KO animals performed equivalent to wild-type C57Bl/6n mice on rotarod and Catwalk motor tasks, and did not display alterations in anxiety-like behavior on open field and elevated zero maze tasks. However, NSG2 KO animals demonstrated enhanced recall in the Morris water maze, accelerated reversal learning in a touch-screen task, and accelerated acquisition and enhanced recall on a Trace Fear Conditioning task. Together, these data point to a specific involvement of NSG2 on multiple types of associative learning, and expand the repertoire of pathways that can be targeted for cognitive enhancement.
Collapse
Affiliation(s)
- Amber J Zimmerman
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
- Present Address: Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Antonio Serrano-Rodriguez
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - Melody Sun
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - Sandy J Wilson
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - David N Linsenbardt
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - Jason P Weick
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA.
| |
Collapse
|
3
|
Zhang H, Li Y, Wang R, Hu X, Wang Z. Neuron-Specific Gene Family Member 1 is a Potential New Therapeutic Target Associated with Immune Cell Infiltration for Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:769-783. [PMID: 39564093 PMCID: PMC11575459 DOI: 10.2147/bctt.s483757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024]
Abstract
Background Breast cancer (BC) is the most common cancer and is highly morphologically and molecularly heterogeneous. Neuron-specific gene family member 1 (NSG1) is a small single-channel transmembrane protein that consists of 185 amino acids and has been reported in a variety of tumours in recent years. However, the role of NSG1 in BC is unclear. Objective This study aimed to explore the role of NSG1 in the pathogenesis and development of BC and its potential as a prognostic marker for BC. Methods This study analysed data from The Cancer Genome Atlas database and the Gene Expression Omnibus database to determine the expression level and prognostic value of NSG1 messenger ribonucleic acid in BC. Using this data, we constructed a clinical risk model. Immunohistochemistry was performed in combination with a clinical cohort of 192 patients with BC to explore the NSG1 protein expression in BC. Enrichment analysis was used to predict the biological function of NSG1 in BC. To analyse the correlation between NSG1 and the BC immune microenvironment, a single-cell analysis of NSG1 expression and cells in BC was performed. Kaplan‒Meier curves and Cox regression analysis were utilised to identify the relationship between the expression of NSG1 protein and clinicopathological features and prognosis. Results Neuron-specific gene family member 1 is highly expressed in patients with early BC, and its expression suggests a good prognosis for patients with BC. Neuron-specific gene family member 1 is involved in the T-cell receptor complex in BC and is associated with CD8 T cells in the BC immune microenvironment and may induce M1 polarisation of macrophages. Conclusion Neuron-specific gene family member 1 is a biomarker of good prognosis in BC. It is associated with the immune microenvironment of BC and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Haoyun Zhang
- Department of Breast Surgery, Affiliated Hospital of Hebei Engineering University, Handan, Hebei, 056000, People's Republic of China
| | - Ying Li
- Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, Handan, Hebei, 056000, People's Republic of China
| | - Ran Wang
- Department of Emergency, Affiliated Hospital of Hebei Engineering University, Handan, Hebei, 056000, People's Republic of China
| | - Xindan Hu
- Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, Handan, Hebei, 056000, People's Republic of China
| | - Zai Wang
- Science and Education Division, Affiliated Hospital of Hebei Engineering University, Handan, Hebei, 056000, People's Republic of China
| |
Collapse
|
4
|
Zimmerman AJ, Serrano-Rodriguez A, Wilson SJ, Linsenbardt DN, Brigman JL, Weick J. Knockout of AMPA receptor binding protein Neuron-Specific Gene 2 (NSG2) enhances associative learning and cognitive flexibility. RESEARCH SQUARE 2024:rs.3.rs-4790348. [PMID: 39257983 PMCID: PMC11384823 DOI: 10.21203/rs.3.rs-4790348/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The vast majority of gene mutations and/or gene knockouts result in either no observable changes, or significant deficits in molecular, cellular, or organismal function. However, in a small number of cases, mutant animal models display enhancements in specific behaviors such as learning and memory. To date, most gene deletions shown to enhance cognitive ability generally affect a limited number of pathways such as NMDA receptor- and translation-dependent plasticity, or GABA receptor- and potassium channel-mediated inhibition. While endolysosomal trafficking of AMPA receptors is a critical mediator of synaptic plasticity, mutations in genes that affect AMPAR trafficking either have no effect or are deleterious for synaptic plasticity, learning and memory. NSG2 is one of the three-member family of Neuron-specific genes (NSG1-3), which have been shown to regulate endolysosomal trafficking of a number of proteins critical for neuronal function, including AMPAR subunits (GluA1-2). Based on these findings and the largely universal expression throughout mammalian brain, we predicted that genetic knockout of NSG2 would result in significant impairments across multiple behavioral modalities including motor, affective, and learning/memory paradigms. However, in the current study we show that loss of NSG2 had highly selective effects on associative learning and memory, leaving motor and affective behaviors intact. For instance, NSG2 KO animals performed equivalent to wild-type C57Bl/6n mice on rotarod and Catwalk motor tasks, and did not display alterations in anxiety-like behavior on open field and elevated zero maze tasks. However, NSG2 KO animals demonstrated enhanced recall in the Morris water maze, accelerated reversal learning in a touch-screen task, and accelerated acquisition and enhanced recall on a Trace Fear Conditioning task. Together, these data point to a specific involvement of NSG2 on multiple types of associative learning, and expand the repertoire of pathways that can be targeted for cognitive enhancement.
Collapse
|
5
|
Overby M, Serrano-Rodriguez A, Dadras S, Christiansen AK, Ozcelik G, Lichtenthaler SF, Weick JP, Müller HK. Neuron-specific gene NSG1 binds to and positively regulates sortilin ectodomain shedding via a metalloproteinase-dependent mechanism. J Biol Chem 2023; 299:105446. [PMID: 37949230 PMCID: PMC10704435 DOI: 10.1016/j.jbc.2023.105446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/15/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
Increasing evidence suggests that aberrant regulation of sortilin ectodomain shedding can contribute to amyloid-β pathology and frontotemporal dementia, although the mechanism by which this occurs has not been elucidated. Here, we probed for novel binding partners of sortilin using multiple and complementary approaches and identified two proteins of the neuron-specific gene (NSG) family, NSG1 and NSG2, that physically interact and colocalize with sortilin. We show both NSG1 and NSG2 induce subcellular redistribution of sortilin to NSG1- and NSG2-enriched compartments. However, using cell surface biotinylation, we found only NSG1 reduced sortilin cell surface expression, which caused significant reductions in uptake of progranulin, a molecular determinant for frontotemporal dementia. In contrast, we demonstrate NSG2 has no effect on sortilin cell surface abundance or progranulin uptake, suggesting specificity for NSG1 in the regulation of sortilin cell surface expression. Using metalloproteinase inhibitors and A disintegrin and metalloproteinase 10 KO cells, we further show that NSG1-dependent reduction of cell surface sortilin occurred via proteolytic processing by A disintegrin and metalloproteinase 10 with a concomitant increase in shedding of sortilin ectodomain to the extracellular space. This represents a novel regulatory mechanism for sortilin ectodomain shedding that is regulated in a neuron-specific manner. Furthermore, this finding has implications for the development of strategies for brain-specific regulation of sortilin and possibly sortilin-driven pathologies.
Collapse
Affiliation(s)
- Malene Overby
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Antonio Serrano-Rodriguez
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Somayeh Dadras
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Ann Kathrine Christiansen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gözde Ozcelik
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der lsar, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der lsar, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jason Porter Weick
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
6
|
Glutamate Receptor Interacting Protein 1 in the Dorsal CA1 Drives Alpha-amino-3-hydroxy-5-methyl-4-Isoxazolepropionic Acid Receptor Endocytosis and Exocytosis Bidirectionally and Mediates Forgetting, Exploratory, and Anxiety-like Behavior. Neuroscience 2022; 498:235-248. [PMID: 35863680 DOI: 10.1016/j.neuroscience.2022.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 01/21/2023]
Abstract
Endocytosis of GluA2-containing alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) in CA1 of the hippocampus regulates forgetting; deficits in forgetting nociceptive memory can induce serious stress disorders. As a transporter of GluA2-containing AMPAR, the functions of glutamate receptor interacting protein 1 (GRIP1) in forgetting and possible stress responses remain unclear. Lentivirus-mediated interference of GRIP1 expression or function in the dorsal CA1 of the hippocampus in mice indicated that GRIP1 overexpression enhanced spatial memory, impaired forgetting in a Barnes maze, and induced anxiety-like behavior in the open field and elevated plus-maze test. In contrast, GRIP1 knockdown impaired learning capacity. Furthermore, inhibition of the PDZ2 and PDZ4/5 domains of GRIP1 and GluA2-dn enhanced learning capacity, whereas GluA2-dn impaired spatial memory; inhibition of the PDZ2 and PDZ4/5 domains of GRIP1 also decreased forgetting capacity to some extent. Importantly, inhibition of both the PDZ2 and PDZ4/5 domains of GRIP1 induced anxiety-like behavior but not GluA2-dn. Furthermore, optogenetic control of both GluA1 and GluA2 insertion into the postsynaptic membrane impaired memory and induced anxiety-like behavior. In vitro experiments showed that GRIP1-ov and -dn, inhibition of PDZ2 and PDZ4/5 domains of GRIP1, and GluA2-dn decreased glycine-induced GluA1 and GluA2 exocytosis; meanwhile, GRIP1-ov and -dn, and interference of PDZ2 and PDZ4/5 domains of GRIP1 blocked AMPA- and NMDA-induced GluA1 and GluA2 endocytosis. Overall, these results suggest that GRIP1 drives AMPA receptor endocytosis and exocytosis bidirectionally; furthermore, GRIP1-induced stabilization of anchoring postsynaptic GluA1 and GluA2 impairs forgetting and induces anxiety-like behavior. GRIP1 may provide a potential therapeutic target in posttraumatic syndromes and anxiety disorders.
Collapse
|
7
|
Yap CC, Winckler B. Spatial regulation of endosomes in growing dendrites. Dev Biol 2022; 486:5-14. [PMID: 35306006 PMCID: PMC10646839 DOI: 10.1016/j.ydbio.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/21/2022] [Accepted: 03/13/2022] [Indexed: 01/19/2023]
Abstract
Many membrane proteins are highly enriched in either dendrites or axons. This non-uniform distribution is a critical feature of neuronal polarity and underlies neuronal function. The molecular mechanisms responsible for polarized distribution of membrane proteins has been studied for some time and many answers have emerged. A less well studied feature of neurons is that organelles are also frequently non-uniformly distributed. For instance, EEA1-positive early endosomes are somatodendritic whereas synaptic vesicles are axonal. In addition, some organelles are present in both axons and dendrites, but not distributed uniformly along the processes. One well known example are lysosomes which are abundant in the soma and proximal dendrite, but sparse in the distal dendrite and the distal axon. The mechanisms that determine the spatial distribution of organelles along dendrites are only starting to be studied. In this review, we will discuss the cell biological mechanisms of how the distribution of diverse sets of endosomes along the proximal-distal axis of dendrites might be regulated. In particular, we will focus on the regulation of bulk homeostatic mechanisms as opposed to local regulation. We posit that immature dendrites regulate organelle motility differently from mature dendrites in order to spatially organize dendrite growth, branching and sculpting.
Collapse
|
8
|
Austin R, Chander P, Zimmerman AJ, Overby M, Digilio L, Yap CC, Linsenbardt DN, Müller HK, Weick JP. Global loss of Neuron-specific gene 1 causes alterations in motor coordination, increased anxiety, and diurnal hyperactivity in male mice. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12816. [PMID: 35577358 PMCID: PMC9262855 DOI: 10.1111/gbb.12816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/26/2022]
Abstract
The Neuron-specific gene family (NSG1-3) consists of small endolysosomal proteins that are critical for trafficking multiple receptors and signaling molecules in neurons. NSG1 has been shown to play a critical role in AMPAR recycling from endosomes to plasma membrane during synaptic plasticity. However, to date nothing is known about whether NSG1 is required for normal behavior at an organismal level. Here we performed a battery of behavioral tests to determine whether loss of NSG1 would affect motor, cognitive, and/or affective behaviors, as well as circadian-related activity. Consistent with unique cerebellar expression of NSG1 among family members, we found that NSG1 was obligatory for motor coordination but not for gross motor function or learning. NSG1 knockout (KO) also altered performance across other behavioral modalities including anxiety-related and diurnal activity paradigms. Surprisingly, NSG1 KO did not cause significant impairments across all tasks within a given modality, but had specific effects within each modality. For instance, we found increases in anxiety-related behaviors in tasks with multiple stressors (e.g., elevation and exposure), but not those with a single main stressor (e.g., exposure). Interestingly, NSG1 KO animals displayed a significant increase in locomotor activity during subjective daytime, suggesting a possible impact on diurnal activity rhythms or vigilance. Surprisingly, loss of NSG1 had no effect on hippocampal-dependent learning despite previous studies showing deficits in CA1 long-term potentiation. Together, these findings do not support a role of NSG1 in hippocampal-dependent learning, but support a role in mediating proper neuronal function across amygdalar and cerebellar circuits.
Collapse
Affiliation(s)
- Roman Austin
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Praveen Chander
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Amber J. Zimmerman
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Malene Overby
- Department of Clinical Medicine, Translational Neuropsychiatry UnitAarhus UniversityAarhus CDenmark
| | - Laura Digilio
- Department of Cell BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Chan Choo Yap
- Department of Cell BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - David N. Linsenbardt
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Heidi Kaastrup Müller
- Department of Clinical Medicine, Translational Neuropsychiatry UnitAarhus UniversityAarhus CDenmark
| | - Jason P. Weick
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| |
Collapse
|
9
|
The Effect of Sleep Deprivation and Subsequent Recovery Period on the Synaptic Proteome of Rat Cerebral Cortex. Mol Neurobiol 2022; 59:1301-1319. [PMID: 34988919 PMCID: PMC8857111 DOI: 10.1007/s12035-021-02699-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/13/2021] [Indexed: 12/31/2022]
Abstract
Sleep deprivation (SD) is commonplace in the modern way of life and has a substantial social, medical, and human cost. Sleep deprivation induces cognitive impairment such as loss of executive attention, working memory decline, poor emotion regulation, increased reaction times, and higher cognitive functions are particularly vulnerable to sleep loss. Furthermore, SD is associated with obesity, diabetes, cardiovascular diseases, cancer, and a vast majority of psychiatric and neurodegenerative disorders are accompanied by sleep disturbances. Despite the widespread scientific interest in the effect of sleep loss on synaptic function, there is a lack of investigation focusing on synaptic transmission on the proteome level. In the present study, we report the effects of SD and recovery period (RP) on the cortical synaptic proteome in rats. Synaptosomes were isolated after 8 h of SD performed by gentle handling and after 16 h of RP. The purity of synaptosome fraction was validated with western blot and electron microscopy, and the protein abundance alterations were analyzed by mass spectrometry. We observed that SD and RP have a wide impact on neurotransmitter-related proteins at both the presynaptic and postsynaptic membranes. The abundance of synaptic proteins has changed to a greater extent in consequence of SD than during RP: we identified 78 proteins with altered abundance after SD and 39 proteins after the course of RP. Levels of most of the altered proteins were upregulated during SD, while RP showed the opposite tendency, and three proteins (Gabbr1, Anks1b, and Decr1) showed abundance changes with opposite direction after SD and RP. The functional cluster analysis revealed that a majority of the altered proteins is related to signal transduction and regulation, synaptic transmission and synaptic assembly, protein and ion transport, and lipid and fatty acid metabolism, while the interaction network analysis revealed several connections between the significantly altered proteins and the molecular processes of synaptic plasticity or sleep. Our proteomic data implies suppression of SNARE-mediated synaptic vesicle exocytosis and impaired endocytic processes after sleep deprivation. Both SD and RP altered GABA neurotransmission and affected protein synthesis, several regulatory processes and signaling pathways, energy homeostatic processes, and metabolic pathways.
Collapse
|
10
|
Radler MR, Suber A, Spiliotis ET. Spatial control of membrane traffic in neuronal dendrites. Mol Cell Neurosci 2020; 105:103492. [PMID: 32294508 PMCID: PMC7317674 DOI: 10.1016/j.mcn.2020.103492] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendrites are highly branched and specialized compartments with distinct structures and secretory organelles (e.g., spines, Golgi outposts), and a unique cytoskeletal organization that includes microtubules of mixed polarity. Dendritic membranes are enriched with proteins, which specialize in the formation and function of the post-synaptic membrane of the neuronal synapse. How these proteins partition preferentially in dendrites, and how they traffic in a manner that is spatiotemporally accurate and regulated by synaptic activity are long-standing questions of neuronal cell biology. Recent studies have shed new insights into the spatial control of dendritic membrane traffic, revealing new classes of proteins (e.g., septins) and cytoskeleton-based mechanisms with dendrite-specific functions. Here, we review these advances by revisiting the fundamental mechanisms that control membrane traffic at the levels of protein sorting and motor-driven transport on microtubules and actin filaments. Overall, dendrites possess unique mechanisms for the spatial control of membrane traffic, which might have specialized and co-evolved with their highly arborized morphology.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Ayana Suber
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Diering GH, Huganir RL. The AMPA Receptor Code of Synaptic Plasticity. Neuron 2019; 100:314-329. [PMID: 30359599 DOI: 10.1016/j.neuron.2018.10.018] [Citation(s) in RCA: 591] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023]
Abstract
Changes in the properties and postsynaptic abundance of AMPA-type glutamate receptors (AMPARs) are major mechanisms underlying various forms of synaptic plasticity, including long-term potentiation (LTP), long-term depression (LTD), and homeostatic scaling. The function and the trafficking of AMPARs to and from synapses is modulated by specific AMPAR GluA1-GluA4 subunits, subunit-specific protein interactors, auxiliary subunits, and posttranslational modifications. Layers of regulation are added to AMPAR tetramers through these different interactions and modifications, increasing the computational power of synapses. Here we review the reliance of synaptic plasticity on AMPAR variants and propose "the AMPAR code" as a conceptual framework. The AMPAR code suggests that AMPAR variants will be predictive of the types and extent of synaptic plasticity that can occur and that a hierarchy exists such that certain AMPARs will be disproportionally recruited to synapses during LTP/homeostatic scaling up, or removed during LTD/homeostatic scaling down.
Collapse
Affiliation(s)
- Graham H Diering
- Department of Cell Biology and Physiology, and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Richard L Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
12
|
Bissen D, Foss F, Acker-Palmer A. AMPA receptors and their minions: auxiliary proteins in AMPA receptor trafficking. Cell Mol Life Sci 2019; 76:2133-2169. [PMID: 30937469 PMCID: PMC6502786 DOI: 10.1007/s00018-019-03068-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/12/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022]
Abstract
To correctly transfer information, neuronal networks need to continuously adjust their synaptic strength to extrinsic stimuli. This ability, termed synaptic plasticity, is at the heart of their function and is, thus, tightly regulated. In glutamatergic neurons, synaptic strength is controlled by the number and function of AMPA receptors at the postsynapse, which mediate most of the fast excitatory transmission in the central nervous system. Their trafficking to, at, and from the synapse, is, therefore, a key mechanism underlying synaptic plasticity. Intensive research over the last 20 years has revealed the increasing importance of interacting proteins, which accompany AMPA receptors throughout their lifetime and help to refine the temporal and spatial modulation of their trafficking and function. In this review, we discuss the current knowledge about the roles of key partners in regulating AMPA receptor trafficking and focus especially on the movement between the intracellular, extrasynaptic, and synaptic pools. We examine their involvement not only in basal synaptic function, but also in Hebbian and homeostatic plasticity. Included in our review are well-established AMPA receptor interactants such as GRIP1 and PICK1, the classical auxiliary subunits TARP and CNIH, and the newest additions to AMPA receptor native complexes.
Collapse
Affiliation(s)
- Diane Bissen
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany
| | - Franziska Foss
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
13
|
Neuron-Specific Gene 2 (NSG2) Encodes an AMPA Receptor Interacting Protein That Modulates Excitatory Neurotransmission. eNeuro 2019; 6:eN-NWR-0292-18. [PMID: 30680309 PMCID: PMC6345199 DOI: 10.1523/eneuro.0292-18.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 01/10/2023] Open
Abstract
Neurons have evolved a number of unique protein-coding genes that regulate trafficking of protein complexes within small organelles throughout dendrites and axons. Neuron-specific gene 2 (NSG2) encodes for one of the most abundant proteins in the nervous system during perinatal development. NSG2 belongs to a family of small neuronal endosomal proteins but its function has remained uncharacterized to date. Here, we show that NSG2 is found in discrete punctae restricted to the somatodendritic arbors of developing mouse and human neurons, and a significant proportion of NSG2 punctae colocalize with postsynaptic HOMER1 and surface-expressed AMPA-type glutamate receptors (AMPARs) at excitatory synapses. Immunoprecipitation revealed that NSG2 physically interacts with both the GluA1 and GluA2 AMPAR subunits in mouse brain. Knock-out of NSG2 in mouse hippocampal neurons selectively impaired the frequency of miniature EPSCs (mEPSCs) and caused alterations in PSD95 expression at postsynaptic densities (PSDs). In contrast, NSG2 overexpression caused a significant increase in the amplitude of mEPSCs as well as GluA2 surface expression. Thus, NSG2 functions as an AMPAR-binding protein that is required for normal synapse formation and/or maintenance, and has unique functions compared with other NSG family members.
Collapse
|
14
|
Parkinson GT, Hanley JG. Mechanisms of AMPA Receptor Endosomal Sorting. Front Mol Neurosci 2018; 11:440. [PMID: 30568574 PMCID: PMC6289981 DOI: 10.3389/fnmol.2018.00440] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022] Open
Abstract
The regulation of synaptic AMPA receptors (AMPARs) is critical for excitatory synaptic transmission, synaptic plasticity and the consequent formation of neural circuits during brain development and their modification during learning and memory processes. The number of synaptic AMPARs is regulated through endocytosis, exocytosis and endosomal sorting that results in recycling back to the plasma membrane or degradation in the lysosome. Hence, endo-lysosomal sorting is vitally important in maintaining AMPAR expression at the synapse, and the dynamic regulation of these trafficking events is a key component of synaptic plasticity. A reduction in synaptic strength such as in long-term depression (LTD) involves AMPAR sorting to lysosomes to reduce synaptic AMPAR number, whereas long-term potentiation (LTP) involves an increase in AMPAR recycling to increase the number of AMPARs at synapses. Here, we review our current understanding of the endosomal trafficking routes taken by AMPARs, and the mechanisms involved in AMPAR endosomal sorting, focussing on the numerous AMPAR associated proteins that have been implicated in this complex process. We also discuss how these events are dysregulated in brain disorders.
Collapse
Affiliation(s)
- Gabrielle T Parkinson
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Jonathan G Hanley
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
15
|
Hanley JG. The Regulation of AMPA Receptor Endocytosis by Dynamic Protein-Protein Interactions. Front Cell Neurosci 2018; 12:362. [PMID: 30364226 PMCID: PMC6193100 DOI: 10.3389/fncel.2018.00362] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/25/2018] [Indexed: 11/13/2022] Open
Abstract
The precise regulation of AMPA receptor (AMPAR) trafficking in neurons is crucial for excitatory neurotransmission, synaptic plasticity and the consequent formation and modification of neural circuits during brain development and learning. Clathrin-mediated endocytosis (CME) is an essential trafficking event for the activity-dependent removal of AMPARs from the neuronal plasma membrane, resulting in a reduction in synaptic strength known as long-term depression (LTD). The regulated AMPAR endocytosis that underlies LTD is caused by specific modes of synaptic activity, most notably stimulation of NMDA receptors (NMDARs) and metabotropic glutamate receptors (mGluRs). Numerous proteins associate with AMPAR subunits, directly or indirectly, to control their trafficking, and therefore the regulation of these protein-protein interactions in response to NMDAR or mGluR signaling is a critical feature of synaptic plasticity. This article reviews the protein-protein interactions that are dynamically regulated during synaptic plasticity to modulate AMPAR endocytosis, focussing on AMPAR binding proteins and proteins that bind the core endocytic machinery. In addition, the mechanisms for the regulation of protein-protein interactions are considered, as well as the functional consequences of these dynamic interactions on AMPAR endocytosis.
Collapse
Affiliation(s)
- Jonathan G Hanley
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
16
|
Moretto E, Passafaro M. Recent Findings on AMPA Receptor Recycling. Front Cell Neurosci 2018; 12:286. [PMID: 30233324 PMCID: PMC6129582 DOI: 10.3389/fncel.2018.00286] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/10/2018] [Indexed: 02/04/2023] Open
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPA-Rs) are tetrameric protein complexes that mediate most of the fast-excitatory transmission in response to the neurotransmitter glutamate in neurons. The abundance of AMPA-Rs at the surface of excitatory synapses establishes the strength of the response to glutamate. It is thus evident that neurons need to tightly regulate this feature, particularly in the context of all synaptic plasticity events, which are considered the biological correlates of higher cognitive functions such as learning and memory. AMPA-R levels at the synapse are regulated by insertion of newly synthesized receptors, lateral diffusion on the plasma membrane and endosomal cycling. The latter is likely the most important especially for synaptic plasticity. This process starts with the endocytosis of the receptor from the cell surface and is followed by either degradation, if the receptor is directed to the lysosomal compartment, or reinsertion at the cell surface through a specialized endosomal compartment called recycling endosomes. Although the basic steps of this process have been discovered, the details and participation of additional regulatory proteins are still being discovered. In this review article, we describe the most recent findings shedding light on this crucial mechanism of synaptic regulation.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche (CNR), Milan, Italy
| | - Maria Passafaro
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche (CNR), Milan, Italy
| |
Collapse
|
17
|
Yap CC, Digilio L, McMahon LP, Garcia ADR, Winckler B. Degradation of dendritic cargos requires Rab7-dependent transport to somatic lysosomes. J Cell Biol 2018; 217:3141-3159. [PMID: 29907658 PMCID: PMC6122995 DOI: 10.1083/jcb.201711039] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/30/2018] [Accepted: 05/31/2018] [Indexed: 12/13/2022] Open
Abstract
Neurons are large and long lived, creating high needs for regulating protein turnover. Disturbances in proteostasis lead to aggregates and cellular stress. We characterized the behavior of the short-lived dendritic membrane proteins Nsg1 and Nsg2 to determine whether these proteins are degraded locally in dendrites or centrally in the soma. We discovered a spatial heterogeneity of endolysosomal compartments in dendrites. Early EEA1-positive and late Rab7-positive endosomes are found throughout dendrites, whereas the density of degradative LAMP1- and cathepsin (Cat) B/D-positive lysosomes decreases steeply past the proximal segment. Unlike in fibroblasts, we found that the majority of dendritic Rab7 late endosomes (LEs) do not contain LAMP1 and that a large proportion of LAMP1 compartments do not contain CatB/D. Second, Rab7 activity is required to mobilize distal predegradative LEs for transport to the soma and terminal degradation. We conclude that the majority of dendritic LAMP1 endosomes are not degradative lysosomes and that terminal degradation of dendritic cargos such as Nsg1, Nsg2, and DNER requires Rab7-dependent transport in LEs to somatic lysosomes.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, VA
| | - Laura Digilio
- Department of Cell Biology, University of Virginia, Charlottesville, VA
| | - Lloyd P McMahon
- Department of Cell Biology, University of Virginia, Charlottesville, VA
| | | | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, VA
| |
Collapse
|
18
|
MAP1B Light Chain Modulates Synaptic Transmission via AMPA Receptor Intracellular Trapping. J Neurosci 2017; 37:9945-9963. [PMID: 28904092 DOI: 10.1523/jneurosci.0505-17.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022] Open
Abstract
The regulated transport of AMPA-type glutamate receptors (AMPARs) to the synaptic membrane is a key mechanism to determine the strength of excitatory synaptic transmission in the brain. In this work, we uncovered a new role for the microtubule-associated protein MAP1B in modulating access of AMPARs to the postsynaptic membrane. Using mice and rats of either sex, we show that MAP1B light chain (LC) accumulates in the somatodendritic compartment of hippocampal neurons, where it forms immobile complexes on microtubules that limit vesicular transport. These complexes restrict AMPAR dendritic mobility, leading to the intracellular trapping of receptors and impairing their access to the dendritic surface and spines. Accordingly, increasing MAP1B-LC expression depresses AMPAR-mediated synaptic transmission. This effect is specific for the GluA2 subunit of the AMPAR and requires glutamate receptor interacting protein 1 (GRIP1) interaction with MAP1B-LC. Therefore, MAP1B-LC represents an alternative link between GRIP1-AMPARs and microtubules that does not result in productive transport, but rather limits AMPAR availability for synaptic insertion, with a direct impact on synaptic transmission.SIGNIFICANCE STATEMENT The ability of neurons to modify their synaptic connections, known as synaptic plasticity, is accepted as the cellular basis for learning and memory. One mechanism for synaptic plasticity is the regulated addition and removal of AMPA-type glutamate receptors (AMPARs) at excitatory synapses. In this study, we found that a microtubule-associated protein, MAP1B light chain (MAP1B-LC), participates in this process. MAP1B-LC forms immobile complexes along dendrites. These complexes limit intracellular vesicular trafficking and trap AMPARs inside the dendritic shaft. In this manner, MAP1B restricts the access of AMPARs to dendritic spines and the postsynaptic membrane, contributing to downregulating synaptic transmission.
Collapse
|
19
|
Yap CC, Digilio L, McMahon L, Winckler B. The endosomal neuronal proteins Nsg1/NEEP21 and Nsg2/P19 are itinerant, not resident proteins of dendritic endosomes. Sci Rep 2017; 7:10481. [PMID: 28874679 PMCID: PMC5585371 DOI: 10.1038/s41598-017-07667-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/29/2017] [Indexed: 12/28/2022] Open
Abstract
Membrane traffic critically regulates most aspects of neuronal function. Neurons express many neuronal-specific proteins that regulate membrane traffic, including the poorly understood small transmembrane proteins neural-specific gene 1 and 2 (Nsg1/NEEP21 and Nsg2/P19). Nsg1 has been implicated in regulating endosomal recycling and sorting of several important neuronal receptors. Nsg2 is largely unstudied. At steady-state, Nsg1 and Nsg2 only partially co-localize with known endosomal compartments, and it was suggested that they mark a neuronal-specific endosome. Since Nsg1 localizes to and functions in the dendritic endosome, we set out to discover how Nsg1 and Nsg2 localization to endosomes is regulated in primary rat hippocampal neurons, using quadruple immunolocalization against endogenous proteins, live imaging of dendritic endosomes, and interference approaches against the endosomal regulators Rab5 and Rab7. In contrast to previous conclusions, we now show that Nsg1 and Nsg2 are not resident endosomal proteins, but traffic rapidly from the cell surface to lysosomes and have a half-life of less than two hours. Their partial co-localization with canonical endosomal markers thus reflects their rapid flux towards degradation rather than specific targeting to a singular compartment. These findings will require rethinking of how this class of endosomal proteins regulates trafficking of much longer-lived receptors.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Laura Digilio
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lloyd McMahon
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
20
|
Shi L, Muthusamy N, Smith D, Bergson C. Dynein binds and stimulates axonal motility of the endosome adaptor and NEEP21 family member, calcyon. Int J Biochem Cell Biol 2017; 90:93-102. [PMID: 28734834 DOI: 10.1016/j.biocel.2017.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 01/31/2023]
Abstract
The neuron-enriched, endosomal protein Calcyon (Caly) regulates endocytosis and vesicle sorting, and is important for synaptic plasticity and brain development. In the current investigation of Caly interacting proteins in brain, the microtubule retrograde motor subunit, cytoplasmic dynein 1 heavy chain (DYNC1H), and microtubule structural proteins, α and β tubulin, were identified as Caly associated proteins by MALDI-ToF/ToF. Direct interaction of the Caly-C terminus with dynein and tubulin was further confirmed in in vitro studies. In Cos-7 cells, mCherry-Caly moved along the microtubule network in organelles largely labeled by the late endosome marker Rab7. Expression of the dynein inhibitor CC1, produced striking alterations in Caly distribution, consistent with retrograde motors playing a prominent role in Caly localization and movement. In axons of cultured adult rat sensory neurons, Caly-positive organelles co-localized with dynein intermediate chain (DYNC1I1-isoform IC-1B) and the dynein regulator, lissencephaly 1 (LIS1), both of which co-precipitated from brain with the Caly C-terminus. Manipulation of dynein function in axons altered the motile properties of Caly indicating that Caly vesicles utilize the retrograde motor. Altogether, the current evidence for association with dynein motors raises the possibility that the endocytic and cargo sorting functions of Caly in neurons could be regulated by interaction with the microtubule transport system.
Collapse
Affiliation(s)
- Liang Shi
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Nagendran Muthusamy
- Department of Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Deanna Smith
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
| | - Clare Bergson
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
21
|
Barford K, Yap CC, Dwyer ND, Winckler B. The related neuronal endosomal proteins NEEP21 (Nsg1) and P19 (Nsg2) have divergent expression profiles in vivo. J Comp Neurol 2017; 525:1861-1878. [PMID: 28299779 DOI: 10.1002/cne.24168] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/28/2016] [Accepted: 11/18/2016] [Indexed: 01/23/2023]
Abstract
Endosomal maturation and transport constitutes a complex trafficking system present in all cell types. Neurons have adapted their endosomal system to meet their unique and complex needs. These adaptations include repurposing existing proteins to diversify endocytosis and trafficking, as well as preferential expression of certain regulators more highly in neurons than other cell types. These neuronal regulators include the family of Neuron-Specific Gene family members (Nsg), NEEP21 (Nsg1), and P19 (Nsg2). NEEP21/Nsg1 plays a role in the trafficking of multiple receptors, including the cell adhesion molecule L1/NgCAM, the neurotransmitter receptor GluA2, and β-APP. Recently, we showed that NEEP2/Nsg1 and P19/Nsg2 are not expressed in all neuronal cell types in vitro. However, it is not known where and when NEEP21/Nsg1 and P19/Nsg2 are expressed in vivo, and whether both proteins are always coexpressed. Here, we show that NEEP21/Nsg1 and P19/Nsg2 are present in both overlapping and distinct cell populations in the hippocampus, neocortex, and cerebellum during development. NEEP21/Nsg1 and P19/Nsg2 levels are highest during embryonic development, and expression persists in the juvenile mouse brain. In particular, a subset of layer V cortical neurons retains relatively high expression of both NEEP21/Nsg1 and P19/Nsg2 at postnatal day 16 as well as in the CA1-3 regions of the hippocampus. In the cerebellum, NEEP21/Nsg1 expression becomes largely restricted to Purkinje neurons in adulthood whereas P19/Nsg2 expression strikingly disappears from the cerebellum with age. This divergent and restricted expression likely reflects differential needs for this class of trafficking regulators in different neurons during different stages of maturation.
Collapse
Affiliation(s)
- Kelly Barford
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Noelle D Dwyer
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
22
|
GRASP1 Regulates Synaptic Plasticity and Learning through Endosomal Recycling of AMPA Receptors. Neuron 2017; 93:1405-1419.e8. [PMID: 28285821 PMCID: PMC5382714 DOI: 10.1016/j.neuron.2017.02.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 12/13/2016] [Accepted: 02/16/2017] [Indexed: 11/23/2022]
Abstract
Learning depends on experience-dependent modification of synaptic efficacy and neuronal connectivity in the brain. We provide direct evidence for physiological roles of the recycling endosome protein GRASP1 in glutamatergic synapse function and animal behavior. Mice lacking GRASP1 showed abnormal excitatory synapse number, synaptic plasticity, and hippocampal-dependent learning and memory due to a failure in learning-induced synaptic AMPAR incorporation. We identified two GRASP1 point mutations from intellectual disability (ID) patients that showed convergent disruptive effects on AMPAR recycling and glutamate uncaging-induced structural and functional plasticity. Wild-type GRASP1, but not ID mutants, rescued spine loss in hippocampal CA1 neurons in Grasp1 knockout mice. Together, these results demonstrate a requirement for normal recycling endosome function in AMPAR-dependent synaptic function and neuronal connectivity in vivo, and suggest a potential role for GRASP1 in the pathophysiology of human cognitive disorders.
Collapse
|
23
|
Han M, Mejias R, Chiu SL, Rose R, Adamczyk A, Huganir R, Wang T. Mice lacking GRIP1/2 show increased social interactions and enhanced phosphorylation at GluA2-S880. Behav Brain Res 2017; 321:176-184. [PMID: 28063882 DOI: 10.1016/j.bbr.2016.12.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/26/2016] [Accepted: 12/29/2016] [Indexed: 01/01/2023]
Abstract
Glutamate receptor interacting proteins 1 and 2 (GRIP1/2) play an important role in regulating synaptic trafficking of AMPA receptor 2/3 (GluA2/3) and synaptic strength. Gain-of-function GRIP1 mutations are implicated in social behavioral deficits in autism. To study mechanisms of Grip1/2-mediated AMPA signaling in the regulation of social behaviors, we performed social behavioral testing on neuron-specific Grip1/2-double knockout (DKO) and wild type (WT) mice that are matched for age, sex, and strain background. We determined the expression profile of key signaling proteins in AMPAR, mGluR, mTOR, and GABA pathways in frontal cortex, striatum, and cerebellum of DKO mice. Compared to WT mice, DKO mice show increased sociability in a modified three-chamber social behavioral test [mean±sem for interaction time in seconds; WT: 44.0±5.0; n=10; DKO: 81.0±9.0; n=9; two factor repeated measures ANOVA: F(1,37)=14.45; p<0.01 and planned t-test; p<0.01] and in a dyadic male-male social interaction test (mean±sem for total time in seconds: sniffing, WT-WT, 18.9±1.1; WT-DKO, 42.5±2.1; t-test: p<0.001; following, WT-WT, 7.7±0.72; WT-DKO,14.4±1.8; t-test: p<0.001). Immunoblot studies identified an increase in phosphorylation at GluA2-Serine 880 (GluA2-pS880) in frontal cortex (mean±sem; WT: 0.69±0.06, n=5; DKO: 0.96±0.06, n=6; t-test; p<0.05) and reduced GABAβ3 expression in striatum (mean±sem; WT: 1.16±0.04, n=4; DKO: 0.95±0.06, n=4; t-test; p<0.05) in DKO mice. GluA2-S880 phosphorylation is known to regulate GluA2synaptic recycling, AMPA signaling strength and plasticity. GABAβ3 has been implicated in the etiology and pathogenesis in autism. These data support an important role of Grip1/2-mediated AMPA signaling in regulating social behaviors and disturbance of glutamate- and GABA-signaling in specialized brain regions in autism-related social behavioral deficits.
Collapse
Affiliation(s)
- Mei Han
- McKusick-Nathans Institute of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Rebeca Mejias
- McKusick-Nathans Institute of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Shu-Ling Chiu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Rebecca Rose
- McKusick-Nathans Institute of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Abby Adamczyk
- McKusick-Nathans Institute of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Richard Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Tao Wang
- McKusick-Nathans Institute of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
24
|
Guo S, Zhu X, Jańczewski D, Lee SSC, He T, Teo SLM, Vancso GJ. Measuring protein isoelectric points by AFM-based force spectroscopy using trace amounts of sample. NATURE NANOTECHNOLOGY 2016; 11:817-23. [PMID: 27454881 DOI: 10.1038/nnano.2016.118] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 06/03/2016] [Indexed: 05/14/2023]
Abstract
Protein charge at various pH and isoelectric point (pI) values is important in understanding protein function. However, often only trace amounts of unknown proteins are available and pI measurements cannot be obtained using conventional methods. Here, we show a method based on the atomic force microscope (AFM) to determine pI using minute quantities of proteins. The protein of interest is immobilized on AFM colloidal probes and the adhesion force of the protein is measured against a positively and a negatively charged substrate made by layer-by-layer deposition of polyelectrolytes. From the AFM force-distance curves, pI values with an estimated accuracy of ±0.25 were obtained for bovine serum albumin, myoglobin, fibrinogen and ribonuclease A over a range of 4.7-9.8. Using this method, we show that the pI of the 'footprint' of the temporary adhesive proteins secreted by the barnacle cyprid larvae of Amphibalanus amphitrite is in the range 9.6-9.7.
Collapse
Affiliation(s)
- Shifeng Guo
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, No. 08-03, Singapore 138634, Singapore
| | - Xiaoying Zhu
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, No. 08-03, Singapore 138634, Singapore
| | - Dominik Jańczewski
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, No. 08-03, Singapore 138634, Singapore
- Laboratory of Technological Processes, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Serina Siew Chen Lee
- Tropical Marine Science Institute, National University of Singapore, 18 Kent Ridge Road, 119227 Singapore, Singapore
| | - Tao He
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, No. 08-03, Singapore 138634, Singapore
| | - Serena Lay Ming Teo
- Tropical Marine Science Institute, National University of Singapore, 18 Kent Ridge Road, 119227 Singapore, Singapore
| | - G Julius Vancso
- Institute of Chemical and Engineering Sciences A*STAR, 1 Pesek Road, Jurong Island, 627833 Singapore, Singapore
- MESA+ Institute for Nanotechnology, Materials Science and Technology of Polymers, University of Twente, PO Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
25
|
Modjeski K, Levy S, Ture S, Field D, Shi G, Ko K, Zhu Q, Morrell C. Glutamate Receptor Interacting Protein 1 Regulates CD4(+) CTLA-4 Expression and Transplant Rejection. Am J Transplant 2016; 16:1383-93. [PMID: 26601915 PMCID: PMC5672809 DOI: 10.1111/ajt.13623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 10/30/2015] [Accepted: 11/08/2015] [Indexed: 01/25/2023]
Abstract
PDZ domains are common 80- to 90-amino-acid regions named after the first three proteins discovered to share these domains: postsynaptic density 95, discs large, and zonula occludens. PDZ domain-containing proteins typically interact with the C-terminus of membrane receptors. Glutamate receptor interacting protein 1 (GRIP1), a seven-PDZ domain protein scaffold, regulates glutamate receptor surface expression and trafficking in neurons. We have found that human and mouse T cells also express GRIP1. T cell-specific GRIP1(-/-) mice >11 weeks old had prolonged cardiac allograft survival. Compared with wild-type T cells, in vitro stimulated GRIP1(-/-) T cells had decreased expression of activation markers and increased apoptotic surface marker expression. Surface expression of the strong T cell inhibitory molecule cytotoxic T lymphocyte antigen-4 (CTLA-4) was increased on GRIP1(-/-) T cells from mice >11 weeks old. CTLA-4 increases with T cell stimulation and its surface expression on GRIP1(-/-) T cells remained high after stimulation was removed, indicating a possible internalization defect in GRIP1-deficient T cells. CTLA-4-blocking antibody treatment following heart transplantation led to complete rejection in T cell GRIP1(-/-) mice, indicating that increased CTLA-4 surface expression contributed to the extended graft survival. Our data indicate that GRIP1 regulates T cell activation by regulating CTLA-4 surface expression.
Collapse
Affiliation(s)
- K.L. Modjeski
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA,Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - S.C. Levy
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - S.K. Ture
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - D.J. Field
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - G. Shi
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - K. Ko
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Q. Zhu
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA,Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - C.N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
26
|
Transferrin Receptor Controls AMPA Receptor Trafficking Efficiency and Synaptic Plasticity. Sci Rep 2016; 6:21019. [PMID: 26880306 PMCID: PMC4754636 DOI: 10.1038/srep21019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/14/2016] [Indexed: 02/05/2023] Open
Abstract
Transferrin receptor (TFR) is an important iron transporter regulating iron homeostasis and has long been used as a marker for clathrin mediated endocytosis. However, little is known about its additional function other than iron transport in the development of central nervous system (CNS). Here we demonstrate that TFR functions as a regulator to control AMPA receptor trafficking efficiency and synaptic plasticity. The conditional knockout (KO) of TFR in neural progenitor cells causes mice to develop progressive epileptic seizure, and dramatically reduces basal synaptic transmission and long-term potentiation (LTP). We further demonstrate that TFR KO remarkably reduces the binding efficiency of GluR2 to AP2 and subsequently decreases AMPA receptor endocytosis and recycling. Thus, our study reveals that TFR functions as a novel regulator to control AMPA trafficking efficiency and synaptic plasticity.
Collapse
|
27
|
Arnal L, Longo G, Stupar P, Castez MF, Cattelan N, Salvarezza RC, Yantorno OM, Kasas S, Vela ME. Localization of adhesins on the surface of a pathogenic bacterial envelope through atomic force microscopy. NANOSCALE 2015; 7:17563-17572. [PMID: 26446736 DOI: 10.1039/c5nr04644k] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Bacterial adhesion is the first and a significant step in establishing infection. This adhesion normally occurs in the presence of flow of fluids. Therefore, bacterial adhesins must be able to provide high strength interactions with their target surface in order to maintain the adhered bacteria under hydromechanical stressing conditions. In the case of B. pertussis, a Gram-negative bacterium responsible for pertussis, a highly contagious human respiratory tract infection, an important protein participating in the adhesion process is a 220 kDa adhesin named filamentous haemagglutinin (FHA), an outer membrane and also secreted protein that contains recognition domains to adhere to ciliated respiratory epithelial cells and macrophages. In this work, we obtained information on the cell-surface localization and distribution of the B. pertussis adhesin FHA using an antibody-functionalized AFM tip. Through the analysis of specific molecular recognition events we built a map of the spatial distribution of the adhesin which revealed a non-homogeneous pattern. Moreover, our experiments showed a force induced reorganization of the adhesin on the surface of the cells, which could explain a reinforced adhesive response under external forces. This single-molecule information contributes to the understanding of basic molecular mechanisms used by bacterial pathogens to cause infectious disease and to gain insights into the structural features by which adhesins can act as force sensors under mechanical shear conditions.
Collapse
Affiliation(s)
- L Arnal
- Centro de Investigación y Desarrollo de Fermentaciones Industriales (CINDEFI-CONICET-CCT La Plata), Facultad de Ciencias Exactas, UNLP. 50 No 227, 1900 La Plata, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ctip2-, Satb2-, Prox1-, and GAD65-Expressing Neurons in Rat Cultures: Preponderance of Single- and Double-Positive Cells, and Cell Type-Specific Expression of Neuron-Specific Gene Family Members, Nsg-1 (NEEP21) and Nsg-2 (P19). PLoS One 2015; 10:e0140010. [PMID: 26465886 PMCID: PMC4605768 DOI: 10.1371/journal.pone.0140010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/31/2015] [Indexed: 11/19/2022] Open
Abstract
The brain consists of many distinct neuronal cell types, but which cell types are present in widely used primary cultures of embryonic rodent brain is often not known. We characterized how abundantly four cell type markers (Ctip2, Satb2, Prox1, GAD65) were represented in cultured rat neurons, how easily neurons expressing different markers can be transfected with commonly used plasmids, and whether neuronal-enriched endosomal proteins Nsg-1 (NEEP21) and Nsg-2 (P19) are ubiquitously expressed in all types of cultured neurons. We found that cultured neurons stably maintain cell type identities that are reflective of cell types in vivo. This includes neurons maintaining simultaneous expression of two transcription factors, such as Ctip2+/Satb2+ or Prox1+/Ctip2+ double-positive cells, which have also been described in vivo. Secondly, we established the superior efficiency of CAG promoters for both Lipofectamine-mediated transfection as well as for electroporation. Thirdly, we discovered that Nsg-1 and Nsg-2 were not expressed equally in all neurons: whereas high levels of both Nsg-1 and Nsg-2 were found in Satb2-, Ctip2-, and GAD65-positive neurons, Prox1-positive neurons in hippocampal cultures expressed low levels of both. Our findings thus highlight the importance of identifying neuronal cell types for doing cell biology in cultured neurons: Keeping track of neuronal cell type might uncover effects in assays that might otherwise be masked by the mixture of responsive and non-responsive neurons in the dish.
Collapse
|
29
|
Activity-dependent synaptic GRIP1 accumulation drives synaptic scaling up in response to action potential blockade. Proc Natl Acad Sci U S A 2015; 112:E3590-9. [PMID: 26109571 DOI: 10.1073/pnas.1510754112] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Synaptic scaling is a form of homeostatic plasticity that stabilizes neuronal firing in response to changes in synapse number and strength. Scaling up in response to action-potential blockade is accomplished through increased synaptic accumulation of GluA2-containing AMPA receptors (AMPAR), but the receptor trafficking steps that drive this process remain largely obscure. Here, we show that the AMPAR-binding protein glutamate receptor-interacting protein-1 (GRIP1) is essential for regulated synaptic AMPAR accumulation during scaling up. Synaptic abundance of GRIP1 was enhanced by activity deprivation, directly increasing synaptic GRIP1 abundance through overexpression increased the amplitude of AMPA miniature excitatory postsynaptic currents (mEPSCs), and shRNA-mediated GRIP1 knockdown prevented scaling up of AMPA mEPSCs. Furthermore, knockdown and replace experiments targeting either GRIP1 or GluA2 revealed that scaling up requires the interaction between GRIP1 and GluA2. Finally, GRIP1 synaptic accumulation during scaling up did not require GluA2 binding. Taken together, our data support a model in which activity-dependent trafficking of GRIP1 to synaptic sites drives the forward trafficking and enhanced synaptic accumulation of GluA2-containing AMPAR during synaptic scaling up.
Collapse
|
30
|
Fbxl10 overexpression in murine hematopoietic stem cells induces leukemia involving metabolic activation and upregulation of Nsg2. Blood 2015; 125:3437-46. [PMID: 25872778 DOI: 10.1182/blood-2014-03-562694] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 03/31/2015] [Indexed: 12/21/2022] Open
Abstract
We previously reported that deficiency for Samd9L, which was cloned as a candidate gene for -7/7q- syndrome, accelerated leukemia cooperatively with enhanced expression of a histone demethylase: F-box and leucine-rich repeat protein 10 (Fbxl10, also known as Jhdm1b, Kdm2b, and Ndy1). To further investigate the role of Fbxl10 in leukemogenesis, we generated transgenic (Tg) mice that overexpress Fbxl10 in hematopoietic stem cells (HSCs). Interestingly, Fbxl10 Tg mice developed myeloid or B-lymphoid leukemia with complete penetrance. HSCs from the Tg mice exhibited an accelerated G0/G1-to-S transition with a normal G0 to G1 entry, resulting in pleiotropic progenitor cell expansion. Fbxl10 Tg HSCs displayed enhanced expression of neuron-specific gene family member 2 (Nsg2), and forced expression of Nsg2 in primary bone marrow cells resulted in expansion of immature cells. In addition, the genes involved in mitochondrial oxidative phosphorylation were markedly enriched in Fbxl10 Tg HSCs, coupled with increased cellular adenosine 5'-triphosphate levels. Moreover, chromatin immunoprecipitation followed by sequencing analysis demonstrated that Fbxl10 directly binds to the regulatory regions of Nsg2 and oxidative phosphorylation genes. These findings define Fbxl10 as a bona fide oncogene, whose deregulated expression contributes to the development of leukemia involving metabolic proliferative advantage and Nsg2-mediated impaired differentiation.
Collapse
|
31
|
Trip6 promotes dendritic morphogenesis through dephosphorylated GRIP1-dependent myosin VI and F-actin organization. J Neurosci 2015; 35:2559-71. [PMID: 25673849 DOI: 10.1523/jneurosci.2125-14.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Thyroid receptor-interacting protein 6 (Trip6), a multifunctional protein belonging to the zyxin family of LIM proteins, is involved in various physiological and pathological processes, including cell migration and tumorigenesis. However, the role of Trip6 in neurons remains unknown. Here, we show that Trip6 is expressed in mouse hippocampal neurons and promotes dendritic morphogenesis. Through interaction with the glutamate receptor-interacting protein 1 (GRIP1) and myosin VI, Trip6 is crucial for the total dendritic length and the number of primary dendrites in cultured hippocampal neurons. Trip6 depletion reduces F-actin content and impairs dendritic morphology, and this phenocopies GRIP1 or myosin VI knockdown. Furthermore, phosphorylation of GRIP1(956T) by AKT1 inhibits the interaction between GRIP1 and myosin VI, but facilitates GRIP1 binding to 14-3-3 protein, which is required for regulating F-actin organization and dendritic morphogenesis. Thus, the Trip6-GRIP1-myosin VI interaction and its regulation on F-actin network play a significant role in dendritic morphogenesis.
Collapse
|
32
|
Muthusamy N, Chen YJ, Yin DM, Mei L, Bergson C. Complementary roles of the neuron-enriched endosomal proteins NEEP21 and calcyon in neuronal vesicle trafficking. J Neurochem 2015; 132:20-31. [PMID: 25376768 DOI: 10.1111/jnc.12989] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/17/2014] [Accepted: 10/23/2014] [Indexed: 01/18/2023]
Abstract
Understanding mechanisms governing the trafficking of transmembrane (TM) cargoes to synapses and other specialized membranes in neurons represents a long-standing challenge in cell biology. Investigation of the neuron-enriched endosomal protein of 21 kDa (NEEP21, or NSG1or P21) and Calcyon (Caly, or NSG3) indicates that the emergence of the NEEP21/Caly/P19 gene family could play a vital role in the success of these mechanisms in vertebrates. The upshot of a sizeable body of work is that the NEEP21 and Caly perform distinct endocytic and recycling functions, which impact (i) α amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptor trafficking at excitatory synapses; (ii) transport to/in neuronal axons; as well as (iii) proteolytic processing of amyloid precursor protein and neuregulin 1, suggesting roles in neuron development, synaptic function, and neurodegeneration. We argue that their distinct effects on cargo endocytosis and recycling depend on interactions with vesicle trafficking and synaptic scaffolding proteins. As they play complementary, but opposing roles in cargo endocytosis, recycling, and degradation, balancing NEEP21 and Caly expression levels or activity could be important for homeostasis in a variety of signaling pathways, and also lead to a novel therapeutic strategy for disorders like Alzheimer's disease and schizophrenia. This review focuses on two closely related, neuron-enriched endosomal proteins: NEEP21 and Calcyon which perform distinct roles in regulating receptor endocytosis, recycling, and degradation. Based on an in-depth examination of the literature, we argue that these two proteins carry out complementary yet sometimes opposing vesicle trafficking functions that impact excitatory transmission, transcytosis, axonal transport, and also proteolytic processing by beta-secretase I (BACE1). Finally, we propose that balancing NEEP21 and Calcyon expression and/or activity could be important for homeostasis in a variety of signaling pathways, and also lead to a novel therapeutic strategy for disorders like Alzheimer's disease and schizophrenia. AMPA = α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; NMDA = N-Methyl-D-aspartate.
Collapse
Affiliation(s)
- Nagendran Muthusamy
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | | | | | | | | |
Collapse
|
33
|
Maturational conversion of dendritic early endosomes and their roles in L1-mediated axon growth. J Neurosci 2015; 34:14633-43. [PMID: 25355216 DOI: 10.1523/jneurosci.1837-14.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The function of endosomes is intricately linked to cellular function in all cell types, including neurons. Intriguingly, neurons express cell type-specific proteins that localize to endosomes, but little is known about how these neuronal proteins interface with canonical endosomes and ubiquitously expressed endosomal components, such as EEA1 (Early Endosomal Antigen 1). NEEP21 (Neuronal Early Endosomal Protein 21 kDa) localizes to somatodendritic endosomes, and downregulation of NEEP21 perturbs the correct trafficking of multiple receptors, including glutamate receptors (GluA2) during LTP and amyloidogenic processing of βAPP. Our own work implicated NEEP21 in correct trafficking of the axonal cell adhesion molecule L1/neuron-glia cell adhesion molecule (NgCAM). NEEP21 dynamically localizes with EEA1-positive early endosomes but is also found in EEA1-negative endosomes. Live imaging reveals that NEEP21-positive, EEA1-negative endosomes arise as a consequence of maturational conversion of EEA1/NEEP21 double-positive endosomes. Interfering with EEA1 function causes missorting of L1/NgCAM, axon outgrowth defects on the L1 substrate, and disturbance of NEEP21 localization. Last, we uncover evidence that functional interference with NEEP21 reduces axon and dendrite growth of primary rat hippocampal neurons on L1 substrate but not on N-cadherin substrate, thus implicating endosomal trafficking through somatodendritic early endosomes in L1-mediated axon growth.
Collapse
|
34
|
Han J, Wu P, Wang F, Chen J. S-palmitoylation regulates AMPA receptors trafficking and function: a novel insight into synaptic regulation and therapeutics. Acta Pharm Sin B 2015; 5:1-7. [PMID: 26579419 PMCID: PMC4629138 DOI: 10.1016/j.apsb.2014.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 12/05/2014] [Accepted: 12/09/2014] [Indexed: 01/11/2023] Open
Abstract
Glutamate acting on AMPA-type ionotropic glutamate receptor (AMPAR) mediates the majority of fast excitatory synaptic transmission in the mammalian central nervous system. Dynamic regulation of AMPAR by post-translational modifications is one of the key elements that allow the nervous system to adapt to environment stimulations. S-palmitoylation, an important lipid modification by post-translational addition of a long-chain fatty acid to a cysteine residue, regulates AMPA receptor trafficking, which dynamically affects multiple fundamental brain functions, such as learning and memory. In vivo, S-palmitoylation is controlled by palmitoyl acyl transferases and palmitoyl thioesterases. In this review, we highlight advances in the mechanisms for dynamic AMPA receptors palmitoylation, and discuss how palmitoylation affects AMPA receptors function at synapses in recent years. Pharmacological regulation of S-palmitoylation may serve as a novel therapeutic strategy for neurobiological diseases.
Collapse
Key Words
- 17-ODYA, 17-octadecynoic acid
- ABE, acyl-biotinyl exchange
- ABP, AMPA receptor binding protein
- AD, Alzheimer׳s disease
- AKAP79/150, A-kinase anchoring protein 79/150
- AMPA receptors
- AMPAR, α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor
- APT1, acyl-protein thioesterase-1
- APT2, acyl-protein thioesterase-2
- CP-AMPARs, Ca2+-permeable AMPARs
- DHHC
- DHHC, aspartate-histidine-histidine-cysteine
- FMRP, fragile X mental retardation protein
- FXS, Fragile X syndrome
- GAP-43, growth associated protein-43
- GRIP, glutamate receptor interacting protein
- LTD, long-term depression
- LTP, long-term potentiation
- PATs, palmitoyl acyl transferases
- PDZ, postsynaptic density-95/discs large/zona occludens-1
- PICK1, protein interacting with C-kinase 1
- PKA, protein kinase A
- PKC, protein kinase C
- PPT1, palmitoyl-protein thioesterase-1
- PSD-95, postsynaptic density-95
- Palmitoylation
- Ras, rat sarcoma
- SNAP-23, soluble N-ethylmaleimide-sensitive fusion protein-attachment protein receptor protein-23
- Trafficking
Collapse
|
35
|
Rubio MD, Drummond JB, Meador-Woodruff JH. Glutamate receptor abnormalities in schizophrenia: implications for innovative treatments. Biomol Ther (Seoul) 2014; 20:1-18. [PMID: 24116269 PMCID: PMC3792192 DOI: 10.4062/biomolther.2012.20.1.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/25/2011] [Indexed: 01/18/2023] Open
Abstract
Schizophrenia is a devastating psychiatric illness that afflicts 1% of the population worldwide, resulting in substantial impact to patients, their families, and health care delivery systems. For many years, schizophrenia has been felt to be associated with dysregulated dopaminergic neurotransmission as a key feature of the pathophysiology of the illness. Although numerous studies point to dopaminergic abnormalities in schizophrenia, dopamine dysfunction cannot completely account for all of the symptoms seen in schizophrenia, and dopamine-based treatments are often inadequate and can be associated with serious side effects. More recently, converging lines of evidence have suggested that there are abnormalities of glutamate transmission in schizophrenia. Glutamatergic neurotransmission involves numerous molecules that facilitate glutamate release, receptor activation, glutamate reuptake, and other synaptic activities. Evidence for glutamatergic abnormalities in schizophrenia primarily has implicated the NMDA and AMPA subtypes of the glutamate receptor. The expression of these receptors and other molecules associated with glutamate neurotransmission has been systematically studied in the brain in schizophrenia. These studies have generally revealed region- and molecule-specific changes in glutamate receptor transcript and protein expression in this illness. Given that glutamatergic neurotransmission has been implicated in the pathophysiology of schizophrenia, recent drug development efforts have targeted the glutamate system. Much effort to date has focused on modulation of the NMDA receptor, although more recently other glutamate receptors and transporters have been the targets of drug development. These efforts have been promising thus far, and ongoing efforts to develop additional drugs that modulate glutamatergic neurotransmission are underway that may hold the potential for novel classes of more effective treatments for this serious psychiatric illness.
Collapse
Affiliation(s)
- Maria D Rubio
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294-0021, USA
| | | | | |
Collapse
|
36
|
Karataeva AR, Klaassen RV, Ströder J, Ruiperez-Alonso M, Hjorth JJJ, van Nierop P, Spijker S, Mansvelder HD, Smit AB. C-terminal interactors of the AMPA receptor auxiliary subunit Shisa9. PLoS One 2014; 9:e87360. [PMID: 24498314 PMCID: PMC3911953 DOI: 10.1371/journal.pone.0087360] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 12/23/2013] [Indexed: 01/01/2023] Open
Abstract
Shisa9 (initially named CKAMP44) has been identified as auxiliary subunit of the AMPA-type glutamate receptors and was shown to modulate its physiological properties. Shisa9 is a type-I transmembrane protein and contains a C-terminal PDZ domain that potentially interacts with cytosolic proteins. In this study, we performed a yeast two-hybrid screening that yielded eight PDZ domain-containing interactors of Shisa9, which were independently validated. The identified interactors are known scaffolding proteins residing in the neuronal postsynaptic density. To test whether C-terminal scaffolding interactions of Shisa9 affect synaptic AMPA receptor function in the hippocampus, we disrupted these interactions using a Shisa9 C-terminal mimetic peptide. In the absence of scaffolding interactions of Shisa9, glutamatergic AMPA receptor-mediated synaptic currents in the lateral perforant path of the mouse hippocampus had a faster decay time, and paired-pulse facilitation was reduced. Furthermore, disruption of the PDZ interactions between Shisa9 and its binding partners affected hippocampal network activity. Taken together, our data identifies novel interaction partners of Shisa9, and shows that the C-terminal interactions of Shisa9 through its PDZ domain interaction motif are important for AMPA receptor synaptic and network functions.
Collapse
Affiliation(s)
- Anna R. Karataeva
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Remco V. Klaassen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Jasper Ströder
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Marta Ruiperez-Alonso
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Johannes J. J. Hjorth
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Pim van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Sabine Spijker
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Huibert D. Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
37
|
Hossain S, Liu HN, Fragoso G, Almazan G. Agonist-induced down-regulation of AMPA receptors in oligodendrocyte progenitors. Neuropharmacology 2014; 79:506-14. [PMID: 24412648 DOI: 10.1016/j.neuropharm.2013.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 12/09/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
Prolonged exposure of oligodendrocyte progenitor cultures to non-toxic concentrations of glutamate receptor agonists for 24 h decreased cellular proliferation mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Since prolonged agonist stimulation can regulate the expression of various families of receptors, we examined this possibility. Pretreatment of progenitor cultures with 100 μM kainic acid (KA) for 1-24 h caused a time-dependent decrease in AMPA receptor activity, determined by agonist-induced (45)Ca(2+) uptake. The maximum effect (70-80% decrease), observed in the 24 h-pretreated cells, was accompanied by a significant reduction in AMPA receptor subunits, as determined by Western blotting. GluR2/3 and GluR4 subunits were the most affected. Receptor down-regulation and (45)Ca(2+) uptake were only partially reversible upon KA removal. Furthermore, 24 h co-treatment of cultures with CNQX blocked the KA-induced decreases in calcium uptake. To address whether calpain, a calcium-activated protease, was implicated in the regulation of the AMPA receptor subunits, cultures were treated with the specific inhibitor PD150606 alone or in combination with KA for 24 h. Calpain inhibition significantly increased GluR1 in both conditions and partly reversed downregulation of GluR4 by KA. Collectively, these results indicate that calpain is not involved in the agonist-induced down-regulation of AMPA receptors subunits 2/3 in oligodendrocyte progenitors, while it downregulates GluR1 and GluR4.
Collapse
Affiliation(s)
- Shireen Hossain
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Hsueh-Ning Liu
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Gabriela Fragoso
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Guillermina Almazan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
38
|
Bassani S, Folci A, Zapata J, Passafaro M. AMPAR trafficking in synapse maturation and plasticity. Cell Mol Life Sci 2013; 70:4411-30. [PMID: 23475111 PMCID: PMC11113961 DOI: 10.1007/s00018-013-1309-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 02/15/2013] [Accepted: 02/18/2013] [Indexed: 12/15/2022]
Abstract
Glutamate ionotropic alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (AMPARs) mediate most fast excitatory synaptic transmission in the central nervous system. The content and composition of AMPARs in postsynaptic membranes (which determine synaptic strength) are dependent on the regulated trafficking of AMPAR subunits in and out of the membranes. AMPAR trafficking is a key mechanism that drives nascent synapse development, and is the main determinant of both Hebbian and homeostatic plasticity in mature synapses. Hebbian plasticity seems to be the biological substrate of at least some forms of learning and memory; while homeostatic plasticity (also known as synaptic scaling) keeps neuronal circuits stable by maintaining changes within a physiological range. In this review, we examine recent findings that provide further understanding of the role of AMPAR trafficking in synapse maturation, Hebbian plasticity, and homeostatic plasticity.
Collapse
Affiliation(s)
- Silvia Bassani
- CNR Institute of Neuroscience, Department of Medical Pharmacology, University of Milan, Milan, Italy
| | - Alessandra Folci
- CNR Institute of Neuroscience, Department of Medical Pharmacology, University of Milan, Milan, Italy
| | - Jonathan Zapata
- CNR Institute of Neuroscience, Department of Medical Pharmacology, University of Milan, Milan, Italy
| | - Maria Passafaro
- CNR Institute of Neuroscience, Department of Medical Pharmacology, University of Milan, Milan, Italy
- Dulbecco Telethon Institute, Rome, Italy
| |
Collapse
|
39
|
Abstract
Among the largest cells in the body, neurons possess an immense surface area and intricate geometry that poses many unique cell biological challenges. This morphological complexity is critical for neural circuit formation and enables neurons to compartmentalize cell-cell communication and local intracellular signalling to a degree that surpasses other cell types. The adaptive plastic properties of neurons, synapses and circuits have been classically studied by measurement of electrophysiological properties, ionic conductances and excitability. Over the last 15 years, the field of synaptic and neural electrophysiology has collided with neuronal cell biology to produce a more integrated understanding of how these remarkable highly differentiated cells utilize common eukaryotic cellular machinery to decode, integrate and propagate signals in the nervous system. The present article gives a very brief and personal overview of the organelles and trafficking machinery of neuronal dendrites and their role in dendritic and synaptic plasticity.
Collapse
Affiliation(s)
- Michael D Ehlers
- *Neuroscience Research Unit, Pfizer Worldwide Research and Development, 700 Main Street, Cambridge, MA 02139, U.S.A
| |
Collapse
|
40
|
Barry J, Gu C. Coupling mechanical forces to electrical signaling: molecular motors and the intracellular transport of ion channels. Neuroscientist 2013; 19:145-59. [PMID: 22910031 PMCID: PMC3625366 DOI: 10.1177/1073858412456088] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proper localization of various ion channels is fundamental to neuronal functions, including postsynaptic potential plasticity, dendritic integration, action potential initiation and propagation, and neurotransmitter release. Microtubule-based forward transport mediated by kinesin motors plays a key role in placing ion channel proteins to correct subcellular compartments. PDZ- and coiled-coil-domain proteins function as adaptor proteins linking ionotropic glutamate and GABA receptors to various kinesin motors, respectively. Recent studies show that several voltage-gated ion channel/transporter proteins directly bind to kinesins during forward transport. Three major regulatory mechanisms underlying intracellular transport of ion channels are also revealed. These studies contribute to understanding how mechanical forces are coupled to electrical signaling and illuminating pathogenic mechanisms in neurodegenerative diseases.
Collapse
Affiliation(s)
- Joshua Barry
- The Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Chen Gu
- The Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
41
|
Abstract
Intracellular membrane trafficking along endocytic and secretory transport pathways plays a critical role in diverse cellular functions including both developmental and pathological processes. Briefly, proteins and lipids destined for transport to distinct locations are collectively assembled into vesicles and delivered to their target site by vesicular fusion. SNARE (soluble N-ethylmaleimide-sensitive factor-attachment protein receptor) proteins are required for these events, during which v-SNAREs (vesicle SNAREs) interact with t-SNAREs (target SNAREs) to allow transfer of cargo from donor vesicle to target membrane. Recently, the t-SNARE family member, syntaxin-6, has been shown to play an important role in the transport of proteins that are key to diverse cellular dynamic processes. In this paper, we briefly discuss the specific role of SNAREs in various mammalian cell types and comprehensively review the various roles of the Golgi- and endosome-localized t-SNARE, syntaxin-6, in membrane trafficking during physiological as well as pathological conditions.
Collapse
|
42
|
Muthusamy N, Faundez V, Bergson C. Calcyon, a mammalian specific NEEP21 family member, interacts with adaptor protein complex 3 (AP-3) and regulates targeting of AP-3 cargoes. J Neurochem 2012; 123:60-72. [PMID: 22650988 DOI: 10.1111/j.1471-4159.2012.07814.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Calcyon is a neural enriched, single transmembrane protein that interacts with clathrin light chain and stimulates clathrin assembly and clathrin-mediated endocytosis. A similar property is shared by the heterotetrameric adaptor protein (AP) complexes AP-1, AP-2, and AP-3 which recruit cargoes for insertion into clathrin coated transport vesicles. Here we report that AP medium (μ) subunits interact with a YXXØ-type tyrosine motif located at residues 133-136 in the cytoplasmic domain of calcyon. Site specific mutagenesis of the critical tyrosine and bulky hydrophobic residues tyrosine 133 and methionine 136 preferentially abrogated binding of the ubiquitous and neuronal isoforms of μ3, and also impacted μ1 and μ2 binding to a lesser degree. The relevance of these interactions was explored in vivo using mice harboring null alleles of calcyon. As seen in the mutagenesis studies, calcyon deletion in mice preferentially altered the subcellular distribution of AP-3 suggesting that calcyon could regulate membrane-bound pools of AP-3 and AP-3 function. To test this hypothesis, we focused on the hilar region of hippocampus, where levels of calcyon, AP-3, and AP-3 cargoes are abundant. We analyzed brain cryosections from control and calcyon null mice for zinc transporter 3 (ZnT3), and phosphatidylinositol-4-kinase type II alpha (PI4KIIα), two well-defined AP-3 cargoes. Confocal microscopy indicated that ZnT3 and PI4KIIα are significantly reduced in the hippocampal mossy fibers of calcyon knock-out brain, a phenotype previously described in AP-3 deficiencies. Altogether, our data suggest that calcyon directly interacts with μ3A and μ3B, and regulates the subcellular distribution of AP-3 and the targeting of AP-3 cargoes.
Collapse
Affiliation(s)
- Nagendran Muthusamy
- Graduate Program in Neuroscience Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | |
Collapse
|
43
|
Chivet M, Hemming F, Pernet-Gallay K, Fraboulet S, Sadoul R. Emerging role of neuronal exosomes in the central nervous system. Front Physiol 2012; 3:145. [PMID: 22654762 PMCID: PMC3361079 DOI: 10.3389/fphys.2012.00145] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 04/30/2012] [Indexed: 12/24/2022] Open
Abstract
Exosomes are small extracellular vesicles, which stem from endosomes fusing with the plasma membrane, and can be recaptured by receiving cells. They contain lipids, proteins, and RNAs able to modify the physiology of receiving cells. Functioning of the brain relies on intercellular communication between neural cells. These communications can modulate the strength of responses at sparse groups of specific synapses, to modulate circuits underlying associations and memory. Expression of new genes must then follow to stabilize the long-term modifications of the synaptic response. Local changes of the physiology of synapses from one neuron driven by another, have so far been explained by classical signal transduction to modulate transcription, translation, and posttranslational modifications. In vitro evidence now demonstrates that exosomes are released by neurons in a way depending on synaptic activity; these exosomes can be retaken by other neurons suggesting a novel way for inter-neuronal communication. The efficacy of inter-neuronal transfer of biochemical information allowed by exosomes would be far superior to that of direct cell-to-cell contacts or secreted soluble factors. Indeed, lipids, proteins, and RNAs contained in exosomes secreted by emitting neurons could directly modify signal transduction and protein expression in receiving cells. Exosomes could thus represent an ideal mechanism for inter-neuronal transfer of information allowing anterograde and retrograde signaling across synapses necessary for plasticity. They might also allow spreading across the nervous system of pathological proteins like PrPsc, APP fragments, phosphorylated Tau, or Alpha-synuclein.
Collapse
Affiliation(s)
- Mathilde Chivet
- U836, Equipe 2, Neurodégénérescence et Plasticité, INSERM Grenoble, France
| | | | | | | | | |
Collapse
|
44
|
Abstract
Endocytosis and endosomal trafficking play a multitude of roles in cellular function beyond regulating entry of essential nutrients. In this review, we discuss the cell biological principles of endosomal trafficking, the neuronal adaptations to endosomal organization, and the role of endosomal trafficking in neural development. In particular, we consider how cell fate decisions, polarity, migration, and axon outgrowth and guidance are influenced by five endosomal tricks: dynamic modulation of receptor levels by endocytosis and recycling, cargo-specific responses via cargo-specific endocytic regulators, cell-type-specific endocytic regulation, ligand-specific endocytic regulation, and endosomal regulation of ligand processing and trafficking.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Neuroscience, University of Virginia, 409 Lane Road, Charlottesville, VA 22908, USA
| | | |
Collapse
|
45
|
Anggono V, Huganir RL. Regulation of AMPA receptor trafficking and synaptic plasticity. Curr Opin Neurobiol 2012; 22:461-9. [PMID: 22217700 DOI: 10.1016/j.conb.2011.12.006] [Citation(s) in RCA: 456] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 12/12/2011] [Accepted: 12/13/2011] [Indexed: 01/15/2023]
Abstract
AMPA receptors (AMPARs) mediate the majority of fast excitatory synaptic transmission in the brain. Dynamic changes in neuronal synaptic efficacy, termed synaptic plasticity, are thought to underlie information coding and storage in learning and memory. One major mechanism that regulates synaptic strength involves the tightly regulated trafficking of AMPARs into and out of synapses. The life cycle of AMPARs from their biosynthesis, membrane trafficking, and synaptic targeting to their degradation are controlled by a series of orchestrated interactions with numerous intracellular regulatory proteins. Here we review recent progress made toward the understanding the regulation of AMPAR trafficking, focusing on the roles of several key intracellular AMPAR interacting proteins.
Collapse
Affiliation(s)
- Victor Anggono
- Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | | |
Collapse
|
46
|
Schmitt A, Leonardi-Essmann F, Durrenberger PF, Wichert SP, Spanagel R, Arzberger T, Kretzschmar H, Zink M, Herrera-Marschitz M, Reynolds R, Rossner MJ, Falkai P, Gebicke-Haerter PJ. Structural synaptic elements are differentially regulated in superior temporal cortex of schizophrenia patients. Eur Arch Psychiatry Clin Neurosci 2012; 262:565-77. [PMID: 22441714 PMCID: PMC3464383 DOI: 10.1007/s00406-012-0306-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/01/2012] [Indexed: 11/26/2022]
Abstract
Inaccurate wiring and synaptic pathology appear to be major hallmarks of schizophrenia. A variety of gene products involved in synaptic neurotransmission and receptor signaling are differentially expressed in brains of schizophrenia patients. However, synaptic pathology may also develop by improper expression of intra- and extra-cellular structural elements weakening synaptic stability. Therefore, we have investigated transcription of these elements in the left superior temporal gyrus of 10 schizophrenia patients and 10 healthy controls by genome-wide microarrays (Illumina). Fourteen up-regulated and 22 downregulated genes encoding structural elements were chosen from the lists of differentially regulated genes for further qRT-PCR analysis. Almost all genes confirmed by this method were downregulated. Their gene products belonged to vesicle-associated proteins, that is, synaptotagmin 6 and syntaxin 12, to cytoskeletal proteins, like myosin 6, pleckstrin, or to proteins of the extracellular matrix, such as collagens, or laminin C3. Our results underline the pivotal roles of structural genes that control formation and stabilization of pre- and post-synaptic elements or influence axon guidance in schizophrenia. The glial origin of collagen or laminin highlights the close interrelationship between neurons and glial cells in establishment and maintenance of synaptic strength and plasticity. It is hypothesized that abnormal expression of these and related genes has a major impact on the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Andrea Schmitt
- Department of Psychiatry and Psychotherapy, University of Göttingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lasiecka ZM, Winckler B. Mechanisms of polarized membrane trafficking in neurons -- focusing in on endosomes. Mol Cell Neurosci 2011; 48:278-87. [PMID: 21762782 PMCID: PMC3205304 DOI: 10.1016/j.mcn.2011.06.013] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 06/21/2011] [Accepted: 06/25/2011] [Indexed: 12/13/2022] Open
Abstract
Neurons are polarized cells that have a complex and unique morphology: long processes (axons and dendrites) extending far from the cell body. In addition, the somatodendritic and axonal domains are further divided into specific subdomains, such as synapses (pre- and postsynaptic specializations), proximal and distal dendrites, axon initial segments, nodes of Ranvier, and axon growth cones. The striking asymmetry and complexity of neuronal cells are necessary for their function in receiving, processing and transferring electrical signals, with each domain playing a precise function in these processes. In order to establish and maintain distinct neuronal domains, mechanisms must exist for protein delivery to specific neuronal compartments, such that each compartment has the correct functional molecular composition. How polarized membrane domains are established and maintained is a long-standing question. Transmembrane proteins, such as receptors and adhesion molecules, can be transported to their proper membrane domains by several pathways. The biosynthetic secretory system delivers newly synthesized transmembrane proteins from the ER via the Golgi and trans-Golgi-network (TGN) to the plasma membrane. In addition, the endosomal system is critically involved in many instances in ensuring proper (re)targeting of membrane components because it can internalize and degrade mislocalized proteins, or recycle proteins from one domain to another. The endosomal system is thus crucial for establishing and maintaining neuronal polarity. In this review, we focus mainly on the intracellular compartments that serve as sorting stations for polarized transport, with particular emphasis on the emerging roles of endosomes.
Collapse
Affiliation(s)
- Zofia M Lasiecka
- Department of Neuroscience, University of Virginia Medical School, 409 Lane Rd. Extension, MR4-6116, Charlottesville, VA 22908, USA
| | | |
Collapse
|
48
|
Winckler B, Yap CC. Endocytosis and endosomes at the crossroads of regulating trafficking of axon outgrowth-modifying receptors. Traffic 2011; 12:1099-108. [PMID: 21535338 PMCID: PMC3155643 DOI: 10.1111/j.1600-0854.2011.01213.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In neurons, many receptors must be localized correctly to axons or dendrites for proper function. During development, receptors for nerve growth and guidance are targeted to axons and localized to growth cones where receptor activation by ligands results in promotion or inhibition of axon growth. Signaling outcomes downstream of ligand binding are determined by the location, levels and residence times of receptors on the neuronal plasma membrane. Therefore, the mechanisms controlling the trafficking of these receptors are crucial to the proper wiring of circuits. Membrane proteins accumulate on the axonal surface by multiple routes, including polarized sorting in the trans Golgi network, sorting in endosomes and removal by endocytosis. Endosomes also play important roles in the signaling pathways for both growth-promoting and -inhibiting molecules: signaling endosomes derived from endocytosis are important for signaling from growth cones to cell bodies. Growth-promoting neurotrophins and growth-inhibiting Nogo-A can use EHD4/Pincher-dependent endocytosis at the growth cone for their respective retrograde signaling. In addition to retrograde transport of endosomes, anterograde transport to axons in endosomes also occurs for several receptors, including the axon outgrowth-promoting cell adhesion molecule L1/NgCAM and TrkA. L1/NgCAM also depends on EHD4/Pincher-dependent endocytosis for its axonal polarization. In this review, we will focus on receptors whose trafficking has been reported to be modulated by the EHD4/Pincher family of endosomal regulators, namely L1/NgCAM, Trk and Nogo-A. We will first summarize the pathways underlying the axonal transport of these proteins and then discuss the potential roles of EHD4/Pincher in mediating their endocytosis.
Collapse
Affiliation(s)
- Bettina Winckler
- University of Virginia Medical School, Department of Neuroscience, MR4-6115, 409 Lane Road Ext. Charlottesville, VA 22936, USA
| | - Chan Choo Yap
- University of Virginia Medical School, Department of Neuroscience, MR4-6115, 409 Lane Road Ext. Charlottesville, VA 22936, USA
| |
Collapse
|
49
|
New insights in endosomal dynamics and AMPA receptor trafficking. Semin Cell Dev Biol 2011; 22:499-505. [PMID: 21843653 DOI: 10.1016/j.semcdb.2011.06.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Revised: 06/20/2011] [Accepted: 06/21/2011] [Indexed: 12/15/2022]
Abstract
The trafficking mechanisms that control the density of synaptic AMPA-type glutamate receptors have received significant attention because of their importance for regulating excitatory synaptic transmission and synaptic plasticity in the hippocampus. AMPA receptors are synthesized in the neuronal cell body and reach their postsynaptic targets after a complex journey involving multiple transport steps along different cytoskeleton structures and through various stages of the endocytic pathway. Dendritic spines are important sites for AMPA receptor trafficking and contain the basic components of endosomal recycling. On induction of synaptic plasticity, internalized AMPA receptors undergo endosomal sorting and cycle through early endosomes and recycling endosomes back to the plasma membrane (model for long-term potentiation) or target for degradation to the lysosomes (model for long-term depression). Exciting new studies now provide insight in actin-mediated processes that controls endosomal tubule formation and receptor sorting. This review describes the path of AMPA receptor internalization up to sites of recycling and summarizes recent studies on actin-mediated endosomal receptor sorting.
Collapse
|
50
|
Hammond JC, McCullumsmith RE, Haroutunian V, Meador-Woodruff JH. Endosomal trafficking of AMPA receptors in frontal cortex of elderly patients with schizophrenia. Schizophr Res 2011; 130:260-5. [PMID: 21576009 PMCID: PMC3139693 DOI: 10.1016/j.schres.2011.04.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 04/25/2011] [Accepted: 04/26/2011] [Indexed: 01/08/2023]
Abstract
Several lines of evidence indicate altered trafficking of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors in schizophrenia. Previous reports have implicated alterations in the endosomal trafficking of AMPA receptors in this illness. We hypothesized that late endosome content of AMPA receptor subunits is altered in schizophrenia. Accordingly, we developed a technique to isolate and measure contents of late endosomes from postmortem human tissue. We found no changes in the expression of the AMPA subunits, GluR1-4, in late endosomes from the dorsolateral prefrontal cortex in schizophrenia. We also hypothesized that proteins involved in the sorting and trafficking of AMPA receptors between endosomal compartments would be altered in schizophrenia. We found no changes in expression of multiple proteins associated with these processes (dynamin3, Arc/ARG3.1, NEEP21, GRASP1, liprin α, and syntaxin13). Together, these data suggest that endosomal trafficking of AMPA receptors in the prefrontal cortex may be largely intact in schizophrenia.
Collapse
Affiliation(s)
- John C Hammond
- Department of Neurobiology, University of Alabama Birmingham, Birmingham, AL 35294-0021, United States.
| | | | | | | |
Collapse
|