1
|
Ma L, Sun D, Wen S, Yuan J, Li J, Tan X, Cao S. PSD-95 Protein: A Promising Therapeutic Target in Chronic Pain. Mol Neurobiol 2025; 62:3361-3375. [PMID: 39285025 DOI: 10.1007/s12035-024-04485-x] [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: 10/04/2023] [Accepted: 09/04/2024] [Indexed: 02/04/2025]
Abstract
Chronic pain, as a social public health problem, has a serious impact on the quality of patients' life. Currently, the main drugs used to treat chronic pain are opioids, antipyretic, and nonsteroidal anti-inflammatory drugs (NSAIDs). But the obvious side effects limit their use, so it is urgent to find new therapeutic targets. Postsynaptic density (PSD)-95 protein plays an important role in the occurrence and development of chronic pain. The over-expression of the PSD-95 protein and its interaction with other proteins are closely related to the chronic pain. Besides, the PSD-95-related drugs that inhibit the expression of PSD-95 as well as the interaction with other protein have been proved to treat chronic pain significantly. In conclusion, although more deep studies are needed in the future, these studies indicate that PSD-95 and the related proteins, such as NMDA receptor (NMDAR) subunit 2B (GluN2B), AMPA receptor (AMPAR), calmodulin-dependent protein kinase II (CaMKII), 5-hydroxytryptamine 2A receptor (5-HT2AR), and neuronal nitric oxide synthase (nNOS), are the promising therapeutic targets for chronic pain.
Collapse
Affiliation(s)
- Lulin Ma
- Department of Pain Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Dongdong Sun
- Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Song Wen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Jie Yuan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Jing Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Xinran Tan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Song Cao
- Department of Pain Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China.
| |
Collapse
|
2
|
Hoisington ZW, Gangal H, Phamluong K, Shukla C, Ehinger Y, Moffat JJ, Homanics GE, Wang J, Ron D. Prosapip1 in the dorsal hippocampus mediates synaptic protein composition, long-term potentiation, and spatial memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.13.597459. [PMID: 38915579 PMCID: PMC11195216 DOI: 10.1101/2024.06.13.597459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Prosapip1 is a brain-specific protein localized to the postsynaptic density, where it promotes dendritic spine maturation in primary hippocampal neurons. However, nothing is known about the role of Prosapip1 in vivo. To examine this, we utilized the Cre-loxP system to develop a Prosapip1 neuronal knockout mouse. We found that Prosapip1 controls the synaptic localization of its binding partner SPAR, along with PSD-95 and the GluN2B subunit of the NMDA receptor (NMDAR) in the dorsal hippocampus (dHP). We next sought to identify the potential contribution of Prosapip1 to the activity and function of the NMDAR and found that Prosapip1 plays an important role in NMDAR-mediated transmission and long-term potentiation (LTP) in the CA1 region of the dHP. As LTP is the cellular hallmark of learning and memory, we examined the consequences of neuronal knockout of Prosapip1 on dHP-dependent memory. We found that global or dHP-specific neuronal knockout of Prosapip1 caused a deficit in learning and memory whereas developmental, locomotor, and anxiety phenotypes were normal. Taken together, Prosapip1 in the dHP promotes the proper localization of synaptic proteins which, in turn, facilitates LTP driving recognition, social, and spatial learning and memory.
Collapse
|
3
|
Petrovskiy DV, Butkova TV, Nikolsky KS, Kopylov AT, Nakhod VI, Kulikova LI, Malsagova KA, Kibrik ND, Rudnev VR, Izotov AA, Kaysheva AL. Extended range proteomic analysis of blood plasma from schizophrenia patients. Front Mol Biosci 2024; 11:1483933. [PMID: 39640846 PMCID: PMC11617367 DOI: 10.3389/fmolb.2024.1483933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction The high prevalence of schizophrenia worldwide makes it necessary to proceed from subjective assessment of patient's clinical symptoms in diagnosis making to searching for circulating blood biomarkers. On the one hand, searching for molecular markers and targets for therapeutics will make it possible to refine and detail the molecular mechanisms of pathology development, while on the other hand, it will offer new opportunities for elaborating novel approaches to disease diagnosis and enhance efficacy and timeliness of drug therapy. Methods In this study, we performed an extended-range proteomic analysis of plasma samples collected from 48 study subjects with confirmed diagnosis of schizophrenia and 50 healthy volunteers. The high-resolution tandem mass spectra recorded in the data-dependent acquisition mode were analyzed using the MaxQuant algorithm for the library of known protein sequences and the PowerNovo algorithm for de novo protein sequencing. Results It was demonstrated that both strategies show similar results for high-abundance proteins (≥1 μg/mL). For mid-abundance (10 ng/mL - 1 μg/mL) and low-abundance (<10 ng/mL) proteins, the results obtained by the two search strategies complement each other. Discussion Group-specific proteins for the samples of schizophrenia patients were identified, presumably being involved in synaptic plasticity, angiogenesis, transcriptional regulation, protein stabilization and degradation.
Collapse
Affiliation(s)
- Denis V. Petrovskiy
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Tatiana V. Butkova
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Kirill S. Nikolsky
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Arthur T. Kopylov
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Valeriya I. Nakhod
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Liudmila I. Kulikova
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Kristina A. Malsagova
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Nikolai D. Kibrik
- Moscow Research Institute of Psychiatry – Branch of the V. Serbsky National Medical Research Centre of Psy-chiatry and Narcology of the Ministry of Health of the Russian Federation, Department of Sexology, Moscow, Russia
| | - Vladimir R. Rudnev
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Alexander A. Izotov
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Anna L. Kaysheva
- Laboratory of Structural Proteomics, Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
4
|
Chae S, Lee HJ, Lee HE, Kim J, Jeong YJ, Lin Y, Kim HY, Leriche G, Ehrlich RS, Lingl SC, Seo MD, Lee YH, Yang J, Kim JI, Hoe HS. The dopamine analogue CA140 alleviates AD pathology, neuroinflammation, and rescues synaptic/cognitive functions by modulating DRD1 signaling or directly binding to Abeta. J Neuroinflammation 2024; 21:200. [PMID: 39129007 PMCID: PMC11317008 DOI: 10.1186/s12974-024-03180-x] [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: 07/10/2023] [Accepted: 07/17/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND We recently reported that the dopamine (DA) analogue CA140 modulates neuroinflammatory responses in lipopolysaccharide-injected wild-type (WT) mice and in 3-month-old 5xFAD mice, a model of Alzheimer's disease (AD). However, the effects of CA140 on Aβ/tau pathology and synaptic/cognitive function and its molecular mechanisms of action are unknown. METHODS To investigate the effects of CA140 on cognitive and synaptic function and AD pathology, 3-month-old WT mice or 8-month-old (aged) 5xFAD mice were injected with vehicle (10% DMSO) or CA140 (30 mg/kg, i.p.) daily for 10, 14, or 17 days. Behavioral tests, ELISA, electrophysiology, RNA sequencing, real-time PCR, Golgi staining, immunofluorescence staining, and western blotting were conducted. RESULTS In aged 5xFAD mice, a model of AD pathology, CA140 treatment significantly reduced Aβ/tau fibrillation, Aβ plaque number, tau hyperphosphorylation, and neuroinflammation by inhibiting NLRP3 activation. In addition, CA140 treatment downregulated the expression of cxcl10, a marker of AD-associated reactive astrocytes (RAs), and c1qa, a marker of the interaction of RAs with disease-associated microglia (DAMs) in 5xFAD mice. CA140 treatment also suppressed the mRNA levels of s100β and cxcl10, markers of AD-associated RAs, in primary astrocytes from 5xFAD mice. In primary microglial cells from 5xFAD mice, CA140 treatment increased the mRNA levels of markers of homeostatic microglia (cx3cr1 and p2ry12) and decreased the mRNA levels of a marker of proliferative region-associated microglia (gpnmb) and a marker of lipid-droplet-accumulating microglia (cln3). Importantly, CA140 treatment rescued scopolamine (SCO)-mediated deficits in long-term memory, dendritic spine number, and LTP impairment. In aged 5xFAD mice, these effects of CA140 treatment on cognitive/synaptic function and AD pathology were regulated by dopamine D1 receptor (DRD1)/Elk1 signaling. In primary hippocampal neurons and WT mice, CA140 treatment promoted long-term memory and dendritic spine formation via effects on DRD1/CaMKIIα and/or ERK signaling. CONCLUSIONS Our results indicate that CA140 improves neuronal/synaptic/cognitive function and ameliorates Aβ/tau pathology and neuroinflammation by modulating DRD1 signaling in primary hippocampal neurons, primary astrocytes/microglia, WT mice, and aged 5xFAD mice.
Collapse
Affiliation(s)
- Sehyun Chae
- Neurovascular Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Hyun-Ju Lee
- Neurodegenerative Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Ha-Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-Gil, Ulsan, 44919, Republic of Korea
| | - Jieun Kim
- Neurodegenerative Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Yoo Joo Jeong
- Neurodegenerative Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Yuxi Lin
- Biopharmaceutical Research Center, Korea Basic Science Institute (KBSI), Ochang, ChungBuk, 28119, Republic of Korea
| | - Hye Yun Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-Gil, Ulsan, 44919, Republic of Korea
| | - Geoffray Leriche
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Rachel S Ehrlich
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Sascha Castro Lingl
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Min-Duk Seo
- College of Pharmacy and Department of Molecular Science and Technology, Ajou University, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Young-Ho Lee
- Biopharmaceutical Research Center, Korea Basic Science Institute (KBSI), Ochang, ChungBuk, 28119, Republic of Korea
- Bio-Analytical Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Gyeonggi , 17546, Republic of Korea
| | - Jerry Yang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA.
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-Gil, Ulsan, 44919, Republic of Korea.
| | - Hyang-Sook Hoe
- Neurodegenerative Unit, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea.
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, 42988, Republic of Korea.
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea.
| |
Collapse
|
5
|
Cherra SJ, Lamb R. Interactions between Ras and Rap signaling pathways during neurodevelopment in health and disease. Front Mol Neurosci 2024; 17:1352731. [PMID: 38463630 PMCID: PMC10920261 DOI: 10.3389/fnmol.2024.1352731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
The Ras family of small GTPases coordinates tissue development by modulating cell proliferation, cell-cell adhesion, and cellular morphology. Perturbations of any of these key steps alter nervous system development and are associated with neurological disorders. While the underlying causes are not known, genetic mutations in Ras and Rap GTPase signaling pathways have been identified in numerous neurodevelopmental disorders, including autism spectrum, neurofibromatosis, intellectual disability, epilepsy, and schizophrenia. Despite diverse clinical presentations, intersections between these two signaling pathways may provide a better understanding of how deviations in neurodevelopment give rise to neurological disorders. In this review, we focus on presynaptic and postsynaptic functions of Ras and Rap GTPases. We highlight various roles of these small GTPases during synapse formation and plasticity. Based on genomic analyses, we discuss how disease-related mutations in Ras and Rap signaling proteins may underlie human disorders. Finally, we discuss how recent observations have identified molecular interactions between these pathways and how these findings may provide insights into the mechanisms that underlie neurodevelopmental disorders.
Collapse
Affiliation(s)
- Salvatore J. Cherra
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, United States
| | | |
Collapse
|
6
|
Abstract
Rett syndrome is a neurodevelopmental disorder caused by loss-of-function mutations in the methyl-CpG binding protein-2 (MeCP2) gene that is characterized by epilepsy, intellectual disability, autistic features, speech deficits, and sleep and breathing abnormalities. Neurologically, patients with all three disorders display microcephaly, aberrant dendritic morphology, reduced spine density, and an imbalance of excitatory/inhibitory signaling. Loss-of-function mutations in the cyclin-dependent kinase-like 5 (CDKL5) and FOXG1 genes also cause similar behavioral and neurobiological defects and were referred to as congenital or variant Rett syndrome. The relatively recent realization that CDKL5 deficiency disorder (CDD), FOXG1 syndrome, and Rett syndrome are distinct neurodevelopmental disorders with some distinctive features have resulted in separate focus being placed on each disorder with the assumption that distinct molecular mechanisms underlie their pathogenesis. However, given that many of the core symptoms and neurological features are shared, it is likely that the disorders share some critical molecular underpinnings. This review discusses the possibility that deregulation of common molecules in neurons and astrocytes plays a central role in key behavioral and neurological abnormalities in all three disorders. These include KCC2, a chloride transporter, vGlut1, a vesicular glutamate transporter, GluD1, an orphan-glutamate receptor subunit, and PSD-95, a postsynaptic scaffolding protein. We propose that reduced expression or activity of KCC2, vGlut1, PSD-95, and AKT, along with increased expression of GluD1, is involved in the excitatory/inhibitory that represents a key aspect in all three disorders. In addition, astrocyte-derived brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), and inflammatory cytokines likely affect the expression and functioning of these molecules resulting in disease-associated abnormalities.
Collapse
Affiliation(s)
- Santosh R D’Mello
- Department of Biological Sciences, Louisiana State University Shreveport, Shreveport, LA 71104, USA
| |
Collapse
|
7
|
Toni M, Arena C, Cioni C, Tedeschi G. Temperature- and chemical-induced neurotoxicity in zebrafish. Front Physiol 2023; 14:1276941. [PMID: 37854466 PMCID: PMC10579595 DOI: 10.3389/fphys.2023.1276941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/22/2023] [Indexed: 10/20/2023] Open
Abstract
Throughout their lives, humans encounter a plethora of substances capable of inducing neurotoxic effects, including drugs, heavy metals and pesticides. Neurotoxicity manifests when exposure to these chemicals disrupts the normal functioning of the nervous system, and some neurotoxic agents have been linked to neurodegenerative pathologies such as Parkinson's and Alzheimer's disease. The growing concern surrounding the neurotoxic impacts of both naturally occurring and man-made toxic substances necessitates the identification of animal models for rapid testing across a wide spectrum of substances and concentrations, and the utilization of tools capable of detecting nervous system alterations spanning from the molecular level up to the behavioural one. Zebrafish (Danio rerio) is gaining prominence in the field of neuroscience due to its versatility. The possibility of analysing all developmental stages (embryo, larva and adult), applying the most common "omics" approaches (transcriptomics, proteomics, lipidomics, etc.) and conducting a wide range of behavioural tests makes zebrafish an excellent model for neurotoxicity studies. This review delves into the main experimental approaches adopted and the main markers analysed in neurotoxicity studies in zebrafish, showing that neurotoxic phenomena can be triggered not only by exposure to chemical substances but also by fluctuations in temperature. The findings presented here serve as a valuable resource for the study of neurotoxicity in zebrafish and define new scenarios in ecotoxicology suggesting that alterations in temperature can synergistically compound the neurotoxic effects of chemical substances, intensifying their detrimental impact on fish populations.
Collapse
Affiliation(s)
- Mattia Toni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Chiara Arena
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Carla Cioni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science (DIVAS), Università Degli Studi di Milano, Milano, Italy
- CRC “Innovation for Well-Being and Environment” (I-WE), Università Degli Studi di Milano, Milano, Italy
| |
Collapse
|
8
|
Ávila-Flores A, Sánchez-Cabezón JJ, Ochoa-Echeverría A, Checa AI, Rosas-García J, Téllez-Araiza M, Casado S, Liébana R, Santos-Mendoza T, Mérida I. Identification of Host PDZ-Based Interactions with the SARS-CoV-2 E Protein in Human Monocytes. Int J Mol Sci 2023; 24:12793. [PMID: 37628973 PMCID: PMC10454406 DOI: 10.3390/ijms241612793] [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: 07/01/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Proteins containing PDZ (post-synaptic density, PSD-95/disc large, Dlg/zonula occludens, ZO-1) domains assemble signaling complexes that orchestrate cell responses. Viral pathogens target host PDZ proteins by coding proteins containing a PDZ-binding motif (PBM). The presence of a PBM in the SARS-CoV-2 E protein contributes to the virus's pathogenicity. SARS-CoV-2 infects epithelia, but also cells from the innate immune response, including monocytes and alveolar macrophages. This process is critical for alterations of the immune response that are related to the deaths caused by SARS-CoV-2. Identification of E-protein targets in immune cells might offer clues to understanding how SARS-CoV-2 alters the immune response. We analyzed the interactome of the SARS-CoV-2 E protein in human monocytes. The E protein was expressed fused to a GFP tag at the amino terminal in THP-1 monocytes, and associated proteins were identified using a proteomic approach. The E-protein interactome provided 372 partners; only 8 of these harbored PDZ domains, including the cell polarity protein ZO-2, the chemoattractant IL-16, and syntenin. We addressed the expression and localization of the identified PDZ proteins along the differentiation of primary and THP-1 monocytes towards macrophages and dendritic cells. Our data highlight the importance of identifying the functions of PDZ proteins in the maintenance of immune fitness and the viral alteration of inflammatory response.
Collapse
Affiliation(s)
- Antonia Ávila-Flores
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Juan José Sánchez-Cabezón
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Ane Ochoa-Echeverría
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Ana I. Checa
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Jorge Rosas-García
- Laboratory of Transcriptomics and Molecular Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-G.); (M.T.-A.); (T.S.-M.)
| | - Mariana Téllez-Araiza
- Laboratory of Transcriptomics and Molecular Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-G.); (M.T.-A.); (T.S.-M.)
| | - Sara Casado
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Rosa Liébana
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| | - Teresa Santos-Mendoza
- Laboratory of Transcriptomics and Molecular Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-G.); (M.T.-A.); (T.S.-M.)
| | - Isabel Mérida
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, 28049 Madrid, Spain; (J.J.S.-C.); (A.O.-E.); (A.I.C.); (S.C.); (R.L.)
| |
Collapse
|
9
|
Park JS, Choe K, Lee HJ, Park TJ, Kim MO. Neuroprotective effects of osmotin in Parkinson's disease-associated pathology via the AdipoR1/MAPK/AMPK/mTOR signaling pathways. J Biomed Sci 2023; 30:66. [PMID: 37568205 PMCID: PMC10422754 DOI: 10.1186/s12929-023-00961-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most frequent age-related neurodegenerative disorder and is characterized by the loss of dopaminergic neurons. Both environmental and genetic aspects are involved in the pathogenesis of PD. Osmotin is a structural and functional homolog of adiponectin, which regulates the phosphorylation of 5' adenosine monophosphate-activated protein kinase (AMPK) via adiponectin receptor 1 (AdipoR1), thus attenuating PD-associated pathology. Therefore, the current study investigated the neuroprotective effects of osmotin using in vitro and in vivo models of PD. METHODS The study used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced and neuron-specific enolase promoter human alpha-synuclein (NSE-hαSyn) transgenic mouse models and 1-methyl-4-phenylpyridinium (MPP+)- or alpha-synuclein A53T-treated cell models. MPTP was injected at a dose of 30 mg/kg/day for five days, and osmotin was injected twice a week at a dose of 15 mg/kg for five weeks. We performed behavioral tests and analyzed the biochemical and molecular changes in the substantia nigra pars compacta (SNpc) and the striatum. RESULTS Based on our study, osmotin mitigated MPTP- and α-synuclein-induced motor dysfunction by upregulating the nuclear receptor-related 1 protein (Nurr1) transcription factor and its downstream markers tyrosine hydroxylase (TH), dopamine transporter (DAT), and vesicular monoamine transporter 2 (VMAT2). From a pathological perspective, osmotin ameliorated neuronal cell death and neuroinflammation by regulating the mitogen-activated protein kinase (MAPK) signaling pathway. Additionally, osmotin alleviated the accumulation of α-synuclein by promoting the AMPK/mammalian target of rapamycin (mTOR) autophagy signaling pathway. Finally, in nonmotor symptoms of PD, such as cognitive deficits, osmotin restored synaptic deficits, thereby improving cognitive impairment in MPTP- and α-synuclein-induced mice. CONCLUSIONS Therefore, our findings indicated that osmotin significantly rescued MPTP/α-synuclein-mediated PD neuropathology. Altogether, these results suggest that osmotin has potential neuroprotective effects in PD neuropathology and may provide opportunities to develop novel therapeutic interventions for the treatment of PD.
Collapse
Affiliation(s)
- Jun Sung Park
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
| | - Kyonghwan Choe
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229ER Maastricht, the Netherlands
| | - Hyeon Jin Lee
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow, G12 0ZD UK
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
- Alz-Dementia Korea Co., Jinju, 52828 Republic of Korea
| |
Collapse
|
10
|
Li Z. The ALS-Related SynGAP1 Pathogenic Variant Causes Dendritic Spine Loss: Potential Mechanisms of Early-Stage ALS Progression. J Neurosci 2023; 43:2819-2821. [PMID: 37076290 PMCID: PMC10124941 DOI: 10.1523/jneurosci.0099-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 04/21/2023] Open
Affiliation(s)
- Zhanxu Li
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
11
|
Qi C, Luo LD, Feng I, Ma S. Molecular mechanisms of synaptogenesis. Front Synaptic Neurosci 2022; 14:939793. [PMID: 36176941 PMCID: PMC9513053 DOI: 10.3389/fnsyn.2022.939793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
Synapses are the basic units for information processing and storage in the nervous system. It is only when the synaptic connection is established, that it becomes meaningful to discuss the structure and function of a circuit. In humans, our unparalleled cognitive abilities are correlated with an increase in the number of synapses. Additionally, genes involved in synaptogenesis are also frequently associated with neurological or psychiatric disorders, suggesting a relationship between synaptogenesis and brain physiology and pathology. Thus, understanding the molecular mechanisms of synaptogenesis is the key to the mystery of circuit assembly and neural computation. Furthermore, it would provide therapeutic insights for the treatment of neurological and psychiatric disorders. Multiple molecular events must be precisely coordinated to generate a synapse. To understand the molecular mechanisms underlying synaptogenesis, we need to know the molecular components of synapses, how these molecular components are held together, and how the molecular networks are refined in response to neural activity to generate new synapses. Thanks to the intensive investigations in this field, our understanding of the process of synaptogenesis has progressed significantly. Here, we will review the molecular mechanisms of synaptogenesis by going over the studies on the identification of molecular components in synapses and their functions in synaptogenesis, how cell adhesion molecules connect these synaptic molecules together, and how neural activity mobilizes these molecules to generate new synapses. Finally, we will summarize the human-specific regulatory mechanisms in synaptogenesis and results from human genetics studies on synaptogenesis and brain disorders.
Collapse
Affiliation(s)
- Cai Qi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Cai Qi,
| | - Li-Da Luo
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, United States
| | - Irena Feng
- Boston University School of Medicine, Boston, MA, United States
| | - Shaojie Ma
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
12
|
Patel SD, Anand D, Motohashi H, Katsuoka F, Yamamoto M, Lachke SA. Deficiency of the bZIP transcription factors Mafg and Mafk causes misexpression of genes in distinct pathways and results in lens embryonic developmental defects. Front Cell Dev Biol 2022; 10:981893. [PMID: 36092713 PMCID: PMC9459095 DOI: 10.3389/fcell.2022.981893] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/28/2022] [Indexed: 01/11/2023] Open
Abstract
Deficiency of the small Maf proteins Mafg and Mafk cause multiple defects, namely, progressive neuronal degeneration, cataract, thrombocytopenia and mid-gestational/perinatal lethality. Previous data shows Mafg -/-:Mafk +/- compound knockout (KO) mice exhibit cataracts age 4-months onward. Strikingly, Mafg -/-:Mafk -/- double KO mice develop lens defects significantly early in life, during embryogenesis, but the pathobiology of these defects is unknown, and is addressed here. At embryonic day (E)16.5, the epithelium of lens in Mafg -/-:Mafk -/- animals appears abnormally multilayered as demonstrated by E-cadherin and nuclear staining. Additionally, Mafg -/-:Mafk -/- lenses exhibit abnormal distribution of F-actin near the "fulcrum" region where epithelial cells undergo apical constriction prior to elongation and reorientation as early differentiating fiber cells. To identify the underlying molecular changes, we performed high-throughput RNA-sequencing of E16.5 Mafg -/-:Mafk -/- lenses and identified a cohort of differentially expressed genes that were further prioritized using stringent filtering criteria and validated by RT-qPCR. Several key factors associated with the cytoskeleton, cell cycle or extracellular matrix (e.g., Cdk1, Cdkn1c, Camsap1, Col3a1, Map3k12, Sipa1l1) were mis-expressed in Mafg -/-:Mafk -/- lenses. Further, the congenital cataract-linked extracellular matrix peroxidase Pxdn was significantly overexpressed in Mafg -/-:Mafk -/- lenses, which may cause abnormal cell morphology. These data also identified the ephrin signaling receptor Epha5 to be reduced in Mafg -/-:Mafk -/- lenses. This likely contributes to the Mafg -/-:Mafk -/- multilayered lens epithelium pathology, as loss of an ephrin ligand, Efna5 (ephrin-A5), causes similar lens defects. Together, these findings uncover a novel early function of Mafg and Mafk in lens development and identify their new downstream regulatory relationships with key cellular factors.
Collapse
Affiliation(s)
- Shaili D. Patel
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Deepti Anand
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Fumiki Katsuoka
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Salil A. Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE, United States,Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, United States,*Correspondence: Salil A. Lachke,
| |
Collapse
|
13
|
Viard J, Loe-Mie Y, Daudin R, Khelfaoui M, Plancon C, Boland A, Tejedor F, Huganir RL, Kim E, Kinoshita M, Liu G, Haucke V, Moncion T, Yu E, Hindie V, Bléhaut H, Mircher C, Herault Y, Deleuze JF, Rain JC, Simonneau M, Lepagnol-Bestel AM. Chr21 protein-protein interactions: enrichment in proteins involved in intellectual disability, autism, and late-onset Alzheimer's disease. Life Sci Alliance 2022; 5:e202101205. [PMID: 35914814 PMCID: PMC9348576 DOI: 10.26508/lsa.202101205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Down syndrome (DS) is caused by human chromosome 21 (HSA21) trisomy. It is characterized by a poorly understood intellectual disability (ID). We studied two mouse models of DS, one with an extra copy of the <i>Dyrk1A</i> gene (189N3) and the other with an extra copy of the mouse Chr16 syntenic region (Dp(16)1Yey). RNA-seq analysis of the transcripts deregulated in the embryonic hippocampus revealed an enrichment in genes associated with chromatin for the 189N3 model, and synapses for the Dp(16)1Yey model. A large-scale yeast two-hybrid screen (82 different screens, including 72 HSA21 baits and 10 rebounds) of a human brain library containing at least 10<sup>7</sup> independent fragments identified 1,949 novel protein-protein interactions. The direct interactors of HSA21 baits and rebounds were significantly enriched in ID-related genes (<i>P</i>-value < 2.29 × 10<sup>-8</sup>). Proximity ligation assays showed that some of the proteins encoded by HSA21 were located at the dendritic spine postsynaptic density, in a protein network at the dendritic spine postsynapse. We located HSA21 DYRK1A and DSCAM, mutations of which increase the risk of autism spectrum disorder (ASD) 20-fold, in this postsynaptic network. We found that an intracellular domain of DSCAM bound either DLGs, which are multimeric scaffolds comprising receptors, ion channels and associated signaling proteins, or DYRK1A. The DYRK1A-DSCAM interaction domain is conserved in <i>Drosophila</i> and humans. The postsynaptic network was found to be enriched in proteins associated with ARC-related synaptic plasticity, ASD, and late-onset Alzheimer's disease. These results highlight links between DS and brain diseases with a complex genetic basis.
Collapse
Affiliation(s)
- Julia Viard
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Yann Loe-Mie
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Rachel Daudin
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Malik Khelfaoui
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Christine Plancon
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Anne Boland
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Francisco Tejedor
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Universidad Miguel Hernandez-Campus de San Juan, San Juan, Spain
| | - Richard L Huganir
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Science, Nagoya University Graduate School of Science, Nagoya, Japan
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz Institut für Molekulare Pharmakologie (FMP) and Freie Universität Berlin, Berlin, Germany
| | | | - Eugene Yu
- Department of Cellular and Molecular Biology, Roswell Park Division of Graduate School, State University of New York at Buffalo, Buffalo, NY, USA
| | | | | | | | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- INSERM, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
- PHENOMIN, Institut Clinique de la Souris, ICS, GIE CERBM, CNRS, INSERM, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Jean-François Deleuze
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | | | - Michel Simonneau
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
- Université Paris-Saclay, CNRS, ENS Paris-Saclay, CentraleSupélec, LuMIn, Gif sur Yvette, France
- Department of Biology, Ecole Normale Supérieure Paris-Saclay Université Paris-Saclay, Gif sur Yvette, France
| | | |
Collapse
|
14
|
Djemil S, Ressel CR, Abdel-Ghani M, Schneeweis AK, Pak DTS. Central Cholinergic Synapse Formation in Optimized Primary Septal-Hippocampal Co-cultures. Cell Mol Neurobiol 2021; 41:1787-1799. [PMID: 32860154 PMCID: PMC7914286 DOI: 10.1007/s10571-020-00948-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/14/2020] [Indexed: 11/29/2022]
Abstract
Septal innervation of basal forebrain cholinergic neurons to the hippocampus is critical for normal learning and memory and is severely degenerated in Alzheimer's disease. To understand the molecular events underlying physiological cholinergic synaptogenesis and remodeling, as well as pathological loss, we developed an optimized primary septal-hippocampal co-culture system. Hippocampal and septal tissue were harvested from embryonic Sprague-Dawley rat brain and cultured together at varying densities, cell ratios, and in the presence of different growth factors. We identified conditions that produced robust septal-hippocampal synapse formation. We used confocal microscopy with primary antibodies and fluorescent ligands to validate that this system was capable of generating developmentally mature cholinergic synapses. Such synapses were comprised of physiological synaptic partners and mimicked the molecular composition of in vivo counterparts. This co-culture system will facilitate the study of the formation, plasticity, and dysfunction of central mammalian cholinergic synapses.
Collapse
Affiliation(s)
- Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Claire R Ressel
- Department of Biology, Georgetown University, Washington, D.C., USA
| | - Mai Abdel-Ghani
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Amanda K Schneeweis
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., USA.
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., USA.
| |
Collapse
|
15
|
Historical perspective and progress on protein ubiquitination at glutamatergic synapses. Neuropharmacology 2021; 196:108690. [PMID: 34197891 DOI: 10.1016/j.neuropharm.2021.108690] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/07/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
Transcription-translation coupling leads to the production of proteins that are key for controlling essential neuronal processes that include neuronal development and changes in synaptic strength. Although these events have been a prevailing theme in neuroscience, the regulation of proteins via posttranslational signaling pathways are equally relevant for these neuronal processes. Ubiquitin is one type of posttranslational modification that covalently attaches to its targets/substrates. Ubiquitination of proteins play a key role in multiple signaling pathways, the predominant being removal of its substrates by a large molecular machine called the proteasome. Here, I review 40 years of progress on ubiquitination in the nervous system at glutamatergic synapses focusing on axon pathfinding, synapse formation, presynaptic release, dendritic spine formation, and regulation of postsynaptic glutamate receptors. Finally, I elucidate emerging themes in ubiquitin biology that may challenge our current understanding of ubiquitin signaling in the nervous system.
Collapse
|
16
|
Rodriguez AR, Anderson ED, O'Neill KM, McEwan PP, Vigilante NF, Kwon M, Akum BF, Stawicki TM, Meaney DF, Firestein BL. Cytosolic PSD-95 interactor alters functional organization of neural circuits and AMPA receptor signaling independent of PSD-95 binding. Netw Neurosci 2021; 5:166-197. [PMID: 33688611 PMCID: PMC7935033 DOI: 10.1162/netn_a_00173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/26/2020] [Indexed: 11/04/2022] Open
Abstract
Cytosolic PSD-95 interactor (cypin) regulates many aspects of neuronal development and function, ranging from dendritogenesis to synaptic protein localization. While it is known that removal of postsynaptic density protein-95 (PSD-95) from the postsynaptic density decreases synaptic N-methyl-D-aspartate (NMDA) receptors and that cypin overexpression protects neurons from NMDA-induced toxicity, little is known about cypin's role in AMPA receptor clustering and function. Experimental work shows that cypin overexpression decreases PSD-95 levels in synaptosomes and the PSD, decreases PSD-95 clusters/μm2, and increases mEPSC frequency. Analysis of microelectrode array (MEA) data demonstrates that cypin or cypinΔPDZ overexpression increases sensitivity to CNQX (cyanquixaline) and AMPA receptor-mediated decreases in spike waveform properties. Network-level analysis of MEA data reveals that cypinΔPDZ overexpression causes networks to be resilient to CNQX-induced changes in local efficiency. Incorporating these findings into a computational model of a neural circuit demonstrates a role for AMPA receptors in cypin-promoted changes to networks and shows that cypin increases firing rate while changing network functional organization, suggesting cypin overexpression facilitates information relay but modifies how information is encoded among brain regions. Our data show that cypin promotes changes to AMPA receptor signaling independent of PSD-95 binding, shaping neural circuits and output to regions beyond the hippocampus.
Collapse
Affiliation(s)
- Ana R Rodriguez
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Erin D Anderson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Kate M O'Neill
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Przemyslaw P McEwan
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | | | - Munjin Kwon
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Barbara F Akum
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Tamara M Stawicki
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
17
|
Menon S, Goldfarb D, Ho CT, Cloer EW, Boyer NP, Hardie C, Bock AJ, Johnson EC, Anil J, Major MB, Gupton SL. The TRIM9/TRIM67 neuronal interactome reveals novel activators of morphogenesis. Mol Biol Cell 2021; 32:314-330. [PMID: 33378226 PMCID: PMC8098814 DOI: 10.1091/mbc.e20-10-0622] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/25/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
TRIM9 and TRIM67 are neuronally enriched E3 ubiquitin ligases essential for appropriate morphogenesis of cortical and hippocampal neurons and fidelitous responses to the axon guidance cue netrin-1. Deletion of murine Trim9 or Trim67 results in neuroanatomical defects and striking behavioral deficits, particularly in spatial learning and memory. TRIM9 and TRIM67 interact with cytoskeletal and exocytic proteins, but the full interactome is not known. Here we performed the unbiased proximity-dependent biotin identification (BioID) approach to define TRIM9 and TRIM67 protein-protein proximity network in developing cortical neurons and identified putative neuronal TRIM interaction partners. Candidates included cytoskeletal regulators, cytosolic protein transporters, exocytosis and endocytosis regulators, and proteins necessary for synaptic regulation. A subset of high-priority candidates was validated, including Myo16, Coro1A, MAP1B, ExoC1, GRIP1, PRG-1, and KIF1A. For a subset of validated candidates, we utilized total internal reflection fluorescence microscopy to demonstrate dynamic colocalization with TRIM proteins at the axonal periphery, including at the tips of filopodia. Further analysis demonstrated that the RNA interference-based knockdown of the unconventional myosin Myo16 in cortical neurons altered growth cone filopodia density and axonal branching patterns in a TRIM9- and netrin-1-dependent manner. Future analysis of other validated candidates will likely identify novel proteins and mechanisms by which TRIM9 and TRIM67 regulate neuronal form and function. [Media: see text].
Collapse
Affiliation(s)
- Shalini Menon
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Dennis Goldfarb
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Chris T. Ho
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Erica W. Cloer
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Nicholas P. Boyer
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Christopher Hardie
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Andrew J. Bock
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Emma C. Johnson
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Joel Anil
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - M. Ben Major
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
18
|
Zhang G, Li J, Zhang J, Liang X, Wang T, Yin S. A high-density SNP-based genetic map and several economic traits-related loci in Pelteobagrus vachelli. BMC Genomics 2020; 21:700. [PMID: 33028208 PMCID: PMC7542894 DOI: 10.1186/s12864-020-07115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 09/29/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND A high-density genetic linkage map is essential for QTL fine mapping, comparative genome analysis, identification of candidate genes and marker-assisted selection in aquaculture species. Pelteobagrus vachelli is a very popular commercial species in Asia. However, some specific characters hindered achievement of the traditional selective breeding based on phenotypes, such as lack of large-scale genomic resource and short of markers tightly associated with growth, sex determination and hypoxia tolerance related traits. RESULTS By making use of 5059 ddRAD markers in P. vachelli, a high-resolution genetic linkage map was successfully constructed. The map' length was 4047.01 cM by using an interval of 0.11 cm, which is an average marker standard. Comparative genome mapping revealed that a high proportion (83.2%) of markers with a one-to-one correspondence were observed between P. vachelli and P. fulvidraco. Based on the genetic map, 8 significant genome-wide QTLs for 4 weight, 1 body proportion, 2 sex determination, and 1 hypoxia tolerance related traits were detected on 4 LGs. Some SNPs from these significant genome-wide QTLs were observably associated with these phenotypic traits in other individuals by Kompetitive Allele Specific PCR. In addition, two candidate genes for weight, Sipa1 and HSD11B2, were differentially expressed between fast-, medium- and slow-growing P. vachelli. Sema7a, associated with hypoxia tolerance, was induced after hypoxia exposure and reoxygenation. CONCLUSIONS We mapped a set of suggestive and significant QTLs as well as candidate genes for 12 growth, 1 sex determination and 1 hypoxia tolerance related traits based on a high-density genetic linkage map by making use of SNP markers for P. fulvidraco. Our results have offered a valuable method about the much more efficient production of all-male, fast growth and hypoxia tolerance P. vachelli for the aquaculture industry.
Collapse
Affiliation(s)
- Guosong Zhang
- College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
- Key laboratory for physiology biochemistry and application, Heze University, Heze, 274015, Shandong, China
| | - Jie Li
- College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Jiajia Zhang
- College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Xia Liang
- Key laboratory for physiology biochemistry and application, Heze University, Heze, 274015, Shandong, China
| | - Tao Wang
- College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
- Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China
| | - Shaowu Yin
- College of Marine Science and Engineering, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China.
- Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, 222005, China.
| |
Collapse
|
19
|
Folci A, Mirabella F, Fossati M. Ubiquitin and Ubiquitin-Like Proteins in the Critical Equilibrium between Synapse Physiology and Intellectual Disability. eNeuro 2020; 7:ENEURO.0137-20.2020. [PMID: 32719102 PMCID: PMC7544190 DOI: 10.1523/eneuro.0137-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/08/2020] [Accepted: 06/17/2020] [Indexed: 01/04/2023] Open
Abstract
Posttranslational modifications (PTMs) represent a dynamic regulatory system that precisely modulates the functional organization of synapses. PTMs consist in target modifications by small chemical moieties or conjugation of lipids, sugars or polypeptides. Among them, ubiquitin and a large family of ubiquitin-like proteins (UBLs) share several features such as the structure of the small protein modifiers, the enzymatic cascades mediating the conjugation process, and the targeted aminoacidic residue. In the brain, ubiquitination and two UBLs, namely sumoylation and the recently discovered neddylation orchestrate fundamental processes including synapse formation, maturation and plasticity, and their alteration is thought to contribute to the development of neurological disorders. Remarkably, emerging evidence suggests that these pathways tightly interplay to modulate the function of several proteins that possess pivotal roles for brain homeostasis as well as failure of this crosstalk seems to be implicated in the development of brain pathologies. In this review, we outline the role of ubiquitination, sumoylation, neddylation, and their functional interplay in synapse physiology and discuss their implication in the molecular pathogenesis of intellectual disability (ID), a neurodevelopmental disorder that is frequently comorbid with a wide spectrum of brain pathologies. Finally, we propose a few outlooks that might contribute to better understand the complexity of these regulatory systems in regard to neuronal circuit pathophysiology.
Collapse
Affiliation(s)
- Alessandra Folci
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano (MI), Italy
| | - Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve 9 Emanuele - Milan, Italy
| | - Matteo Fossati
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano (MI), Italy
- CNR-Institute of Neuroscience, via Manzoni 56, 20089, Rozzano (MI), Italy
| |
Collapse
|
20
|
Pasciuto E, Burton OT, Roca CP, Lagou V, Rajan WD, Theys T, Mancuso R, Tito RY, Kouser L, Callaerts-Vegh Z, de la Fuente AG, Prezzemolo T, Mascali LG, Brajic A, Whyte CE, Yshii L, Martinez-Muriana A, Naughton M, Young A, Moudra A, Lemaitre P, Poovathingal S, Raes J, De Strooper B, Fitzgerald DC, Dooley J, Liston A. Microglia Require CD4 T Cells to Complete the Fetal-to-Adult Transition. Cell 2020; 182:625-640.e24. [PMID: 32702313 PMCID: PMC7427333 DOI: 10.1016/j.cell.2020.06.026] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 03/05/2020] [Accepted: 06/16/2020] [Indexed: 01/01/2023]
Abstract
The brain is a site of relative immune privilege. Although CD4 T cells have been reported in the central nervous system, their presence in the healthy brain remains controversial, and their function remains largely unknown. We used a combination of imaging, single cell, and surgical approaches to identify a CD69+ CD4 T cell population in both the mouse and human brain, distinct from circulating CD4 T cells. The brain-resident population was derived through in situ differentiation from activated circulatory cells and was shaped by self-antigen and the peripheral microbiome. Single-cell sequencing revealed that in the absence of murine CD4 T cells, resident microglia remained suspended between the fetal and adult states. This maturation defect resulted in excess immature neuronal synapses and behavioral abnormalities. These results illuminate a role for CD4 T cells in brain development and a potential interconnected dynamic between the evolution of the immunological and neurological systems. Video Abstract
Residential CD4 T cells are present in the healthy mouse and human brain Brain residency is a transient program initiated in situ and lasting weeks CD4 T cell entry around birth drives a transcriptional maturation step in microglia Absence of CD4 T cells results in defective synaptic pruning and behavior
Collapse
Affiliation(s)
- Emanuela Pasciuto
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Oliver T Burton
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Carlos P Roca
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Vasiliki Lagou
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Wenson D Rajan
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Tom Theys
- Department of Neurosurgery, UZ Leuven, Leuven 3000, Belgium
| | - Renzo Mancuso
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Neurosciences, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Raul Y Tito
- Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium; VIB-KU Leuven Center for Microbiology, VIB, Leuven 3000, Belgium
| | - Lubna Kouser
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK
| | | | - Alerie G de la Fuente
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Teresa Prezzemolo
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Loriana G Mascali
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Aleksandra Brajic
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Carly E Whyte
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Lidia Yshii
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Anna Martinez-Muriana
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Neurosciences, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Michelle Naughton
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Andrew Young
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Alena Moudra
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Pierre Lemaitre
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | | | - Jeroen Raes
- Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium; VIB-KU Leuven Center for Microbiology, VIB, Leuven 3000, Belgium
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Neurosciences, KU Leuven-University of Leuven, Leuven 3000, Belgium; Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Denise C Fitzgerald
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast BT7 1NN, UK
| | - James Dooley
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Adrian Liston
- VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium; Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven 3000, Belgium; Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK.
| |
Collapse
|
21
|
Transcriptome analysis of the brain provides insights into the regulatory mechanism for Coilia nasus migration. BMC Genomics 2020; 21:410. [PMID: 32552858 PMCID: PMC7302372 DOI: 10.1186/s12864-020-06816-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/09/2020] [Indexed: 11/10/2022] Open
Abstract
Background Coilia nasus (C. nasus) is an important anadromous fish species that resides in the Yangtze River of China, and has high ecological and economical value. However, wild resources have suffered from a serious reduction in population, attributed to the over-construction of water conservancy projects, overfishing, and environmental pollution. The Ministry of Agriculture and Rural Affairs of the People’s Republic of China has issued a notice banning the commercial fishing of wild C. nasus in the Yangtze River. Wild C. nasus populations urgently need to recover. A better understanding of C. nasus migration patterns is necessary to maximize the efficiency of conservation efforts. Juvenile C. nasus experience a simultaneous effect of increasing salinity and cold stress during seaward migration, and the brain plays a comprehensive regulatory role during this process. Therefore, to explore the early seaward migration regulation mechanism of juvenile C. nasus, we performed a comparative transcriptome analysis on the brain of juvenile C. nasus under salinity and cold stress simultaneously. Results Relevant neurotransmitters, receptors, and regulatory proteins from three categories of regulatory pathway play synergistic regulatory roles during the migration process: neuronal signaling, the sensory system, and environmental adaptation. The significant differential expression of growth-related hormones, thyroid receptors, haptoglobin, and prolactin receptors was similar to the results of relevant research on salmonids and steelhead trout. Conclusions This study revealed a regulatory network that the brain of juvenile C. nasus constructs during migration, thereby providing basic knowledge on further studies could build on. This study also revealed key regulatory genes similar to salmonids and steelhead trout, thus, this study will lay a theoretical foundation for further study on migration regulation mechanism of anadromous fish species.
Collapse
|
22
|
Kim Y, Jang YN, Kim JY, Kim N, Noh S, Kim H, Queenan BN, Bellmore R, Mun JY, Park H, Rah JC, Pak DTS, Lee KJ. Microtubule-associated protein 2 mediates induction of long-term potentiation in hippocampal neurons. FASEB J 2020; 34:6965-6983. [PMID: 32237183 DOI: 10.1096/fj.201902122rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/19/2022]
Abstract
Microtubule-associated protein (MAP) 2 has been perceived as a static cytoskeletal protein enriched in neuronal dendritic shafts. Emerging evidence indicates dynamic functions for various MAPs in activity-dependent synaptic plasticity. However, it is unclear how MAP2 is associated with synaptic plasticity mechanisms. Here, we demonstrate that specific silencing of high-molecular-weight MAP2 in vivo abolished induction of long-term potentiation (LTP) in the Schaffer collateral pathway of CA1 pyramidal neurons and in vitro blocked LTP-induced surface delivery of AMPA receptors and spine enlargement. In mature hippocampal neurons, we observed rapid translocation of a subpopulation of MAP2, present in dendritic shafts, to spines following LTP stimulation. Time-lapse confocal imaging showed that spine translocation of MAP2 was coupled with LTP-induced spine enlargement. Consistently, immunogold electron microscopy revealed that LTP stimulation of the Schaffer collateral pathway promoted MAP2 labeling in spine heads of CA1 neurons. This translocation depended on NMDA receptor activation and Ras-MAPK signaling. Furthermore, LTP stimulation led to an increase in surface-expressed AMPA receptors specifically in the neurons with MAP2 spine translocation. Altogether, this study indicates a novel role for MAP2 in LTP mechanisms and suggests that MAP2 participates in activity-dependent synaptic plasticity in mature hippocampal networks.
Collapse
Affiliation(s)
- Yoonju Kim
- Neural Circuits Research Group, Korea Basic Science Research Institute (KBRI), Daegu, Republic of Korea
| | - You-Na Jang
- Neural Circuits Research Group, Korea Basic Science Research Institute (KBRI), Daegu, Republic of Korea
| | - Ji-Young Kim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Nari Kim
- Center for Cortical Processing, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Seulgi Noh
- Neural Circuits Research Group, Korea Basic Science Research Institute (KBRI), Daegu, Republic of Korea
| | - Hyeyeon Kim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Bridget N Queenan
- Department of Pharmacology and Physiology, Interdisciplinary Program of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Ryan Bellmore
- Department of Pharmacology and Physiology, Interdisciplinary Program of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Ji Young Mun
- Neural Circuits Research Group, Korea Basic Science Research Institute (KBRI), Daegu, Republic of Korea
| | - Hyungju Park
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, Republic of Korea
| | - Jong Cheol Rah
- Center for Cortical Processing, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, Republic of Korea
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Interdisciplinary Program of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Kea Joo Lee
- Neural Circuits Research Group, Korea Basic Science Research Institute (KBRI), Daegu, Republic of Korea.,Center for Cortical Processing, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, Republic of Korea
| |
Collapse
|
23
|
Yang N, Zhang Y, Wang JT, Chen C, Song Y, Liang JM, Ma DH, Zhang YF. Effects of Dexamethasone on Remodeling of the Hippocampal Synaptic Filamentous Actin Cytoskeleton in a Model of Pilocarpine-induced Status Epilepticus. Int J Med Sci 2020; 17:1683-1691. [PMID: 32714071 PMCID: PMC7378655 DOI: 10.7150/ijms.44927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/18/2020] [Indexed: 12/28/2022] Open
Abstract
The filamentous actin (F-actin) cytoskeleton is progressively damaged after status epilepticus (SE), which is related to delayed neuronal death, aberrant recurrent circuits and epileptogenesis. Glucocorticoids regulate dendritic spine remodeling by acting on glucocorticoid receptors and the dynamics of the F-actin cytoskeleton. Our previous study showed that administration of dexamethasone (DEX) in the latent period of the pilocarpine epileptic model reduces damage to the hippocampal filamentous actin cytoskeleton and the loss of hippocampal neurons and aids in maintaining the synaptic structures, but it is not sufficient to stop epileptogenesis. In this work, we focused on the role of glucocorticoids in regulating the hippocampal F-actin cytoskeleton during SE. We examined the abundance of synaptic F-actin, analyzed the hippocampal F-actin/G-actin (F/G) ratio and pCofilin, and evaluated the number of hippocampal neurons and pre/postsynaptic markers in pilocarpine-induced status epilepticus mice with or without administration of dexamethasone (DEX). We found that the latency of Stage 3 seizures increased, the mortality decreased, the damage to the synaptic F-actin cytoskeleton in the hippocampal subfields was significantly attenuated, and a greater number of postsynaptic structures were retained in the hippocampal subfields after treatment with DEX. These results indicate that treatment with dexamethasone stabilizes the synaptic F-actin cytoskeleton and reduces the damage to the brain due to SE. This approach is expected to be beneficial in alleviating delayed neuron damage and the process of epileptogenesis.
Collapse
Affiliation(s)
- Nuo Yang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China.,Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Yan Zhang
- College of Life Sciences, Jilin University; Jilin Province, 130021, PR China
| | - Jiang-Tao Wang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Chen Chen
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Yan Song
- Nursing College, Beihua University, 3999 Huashan Road, Jilin 132013, PR China
| | - Jian-Min Liang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Di-Hui Ma
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Yan-Feng Zhang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| |
Collapse
|
24
|
Djemil S, Chen X, Zhang Z, Lee J, Rauf M, Pak DTS, Dzakpasu R. Activation of nicotinic acetylcholine receptors induces potentiation and synchronization within in vitro hippocampal networks. J Neurochem 2019; 153:468-484. [PMID: 31821553 DOI: 10.1111/jnc.14938] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/08/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are known to play a role in cognitive functions of the hippocampus, such as memory consolidation. Given that they conduct Ca2+ and are capable of regulating the release of glutamate and γ-aminobutyric acid (GABA) within the hippocampus, thereby shifting the excitatory-inhibitory ratio, we hypothesized that the activation of nAChRs will result in the potentiation of hippocampal networks and alter synchronization. We used nicotine as a tool to investigate the impact of activation of nAChRs on neuronal network dynamics in primary embryonic rat hippocampal cultures prepared from timed-pregnant Sprague-Dawley rats. We perturbed cultured hippocampal networks with increasing concentrations of bath-applied nicotine and performed network extracellular recordings of action potentials using a microelectrode array. We found that nicotine modulated network dynamics in a concentration-dependent manner; it enhanced firing of action potentials as well as facilitated bursting activity. In addition, we used pharmacological agents to determine the contributions of discrete nAChR subtypes to the observed network dynamics. We found that β4-containing nAChRs are necessary for the observed increases in spiking, bursting, and synchrony, while the activation of α7 nAChRs augments nicotine-mediated network potentiation but is not necessary for its manifestation. We also observed that antagonists of N-methyl-D-aspartate receptors (NMDARs) and group I metabotropic glutamate receptors (mGluRs) partially blocked the effects of nicotine. Furthermore, nicotine exposure promoted autophosphorylation of Ca2+ /calmodulin-dependent kinase II (CaMKII) and serine 831 phosphorylation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunit GluA1. These results suggest that nicotinic receptors induce potentiation and synchronization of hippocampal networks and glutamatergic synaptic transmission. Findings from this work highlight the impact of cholinergic signaling in generating network-wide potentiation in the form of enhanced spiking and bursting dynamics that coincide with molecular correlates of memory such as increased phosphorylation of CaMKII and GluA1. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Xin Chen
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Ziyue Zhang
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Jisoo Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Mikael Rauf
- Department of Human Science, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Rhonda Dzakpasu
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Department of Physics, Georgetown University, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
25
|
Glucagon-like peptide-1 suppresses neuroinflammation and improves neural structure. Pharmacol Res 2019; 152:104615. [PMID: 31881271 DOI: 10.1016/j.phrs.2019.104615] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is a hormone mainly secreted from enteroendocrine L cells. GLP-1 and its receptor are also expressed in the brain. GLP-1 signaling has pivotal roles in regulating neuroinflammation and memory function, but it is unclear how GLP-1 improves memory function by regulating neuroinflammation. Here, we demonstrated that GLP-1 enhances neural structure by inhibiting lipopolysaccharide (LPS)-induced inflammation in microglia with the effects of GLP-1 itself on neurons. Inflammatory secretions of BV-2 microglia by LPS aggravated mitochondrial function and cell survival, as well as neural structure in Neuro-2a neurons. In inflammatory condition, GLP-1 suppressed the secretion of tumor necrosis factor-alpha (TNF-α)-associated cytokines and chemokines in BV-2 microglia and ultimately enhanced neurite complexity (neurite length, number of neurites from soma, and secondary branches) in Neuro-2a neurons. We confirmed that GLP-1 improves neurite complexity, dendritic spine morphogenesis, and spine development in TNF-α-treated primary cortical neurons based on altered expression levels of the factors related to neurite growth and spine morphology. Given that our data that GLP-1 itself enhances neurite complexity and spine morphology in neurons, we suggest that GLP-1 has a therapeutic potential in central nervous system diseases.
Collapse
|
26
|
Khan S, Lu X, Huang Q, Tang J, Weng J, Yang Z, Lv M, Xu X, Xia F, Zhang M, Li Y, Liu S, Leng G, Spitzer N, Du J, Chen X. IGFBP2 Plays an Essential Role in Cognitive Development during Early Life. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901152. [PMID: 31832311 PMCID: PMC6891907 DOI: 10.1002/advs.201901152] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/02/2019] [Indexed: 06/10/2023]
Abstract
Identifying the mechanisms underlying cognitive development in early life is a critical objective. The expression of insulin-like growth factor binding protein 2 (IGFBP2) in the hippocampus increases during neonatal development and is associated with learning and memory, but a causal connection has not been established. Here, it is reported that neurons and astrocytes expressing IGFBP2 are distributed throughout the hippocampus. IGFBP2 enhances excitatory inputs onto CA1 pyramidal neurons, facilitating intrinsic excitability and spike transmission, and regulates plasticity at excitatory synapses in a cell-type specific manner. It facilitates long-term potentiation (LTP) by enhancing N-methyl-d-aspartate (NMDA) receptor-dependent excitatory postsynaptic current (EPSC), and enhances neurite proliferation and elongation. Knockout of igfbp2 reduces the numbers of pyramidal cells and interneurons, impairs LTP and cognitive performance, and reduces tonic excitation of pyramidal neurons that are all rescued by IGFBP2. The results provide insight into the requirement for IGFBP2 in cognition in early life.
Collapse
Affiliation(s)
- Shumsuzzaman Khan
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
- Present address:
Case Western Reserve UniversityClevelandOhioUSA
| | - Xinjiang Lu
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Qingyao Huang
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Jiawei Tang
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Jian Weng
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Zhi Yang
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Minchao Lv
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Xiaokang Xu
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Fangyuan Xia
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Mengchen Zhang
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Yi Li
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Shuangshuang Liu
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Gareth Leng
- Experimental PhysiologyUniversity of EdinburghEdinburghEH8 9XDUK
| | | | - Jizeng Du
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Xuequn Chen
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| |
Collapse
|
27
|
Chidambaram SB, Rathipriya AG, Bolla SR, Bhat A, Ray B, Mahalakshmi AM, Manivasagam T, Thenmozhi AJ, Essa MM, Guillemin GJ, Chandra R, Sakharkar MK. Dendritic spines: Revisiting the physiological role. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:161-193. [PMID: 30654089 DOI: 10.1016/j.pnpbp.2019.01.005] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 01/04/2019] [Accepted: 01/12/2019] [Indexed: 12/11/2022]
Abstract
Dendritic spines are small, thin, specialized protrusions from neuronal dendrites, primarily localized in the excitatory synapses. Sophisticated imaging techniques revealed that dendritic spines are complex structures consisting of a dense network of cytoskeletal, transmembrane and scaffolding molecules, and numerous surface receptors. Molecular signaling pathways, mainly Rho and Ras family small GTPases pathways that converge on actin cytoskeleton, regulate the spine morphology and dynamics bi-directionally during synaptic activity. During synaptic plasticity the number and shapes of dendritic spines undergo radical reorganizations. Long-term potentiation (LTP) induction promote spine head enlargement and the formation and stabilization of new spines. Long-term depression (LTD) results in their shrinkage and retraction. Reports indicate increased spine density in the pyramidal neurons of autism and Fragile X syndrome patients and reduced density in the temporal gyrus loci of schizophrenic patients. Post-mortem reports of Alzheimer's brains showed reduced spine number in the hippocampus and cortex. This review highlights the spine morphogenesis process, the activity-dependent structural plasticity and mechanisms by which synaptic activity sculpts the dendritic spines, the structural and functional changes in spines during learning and memory using LTP and LTD processes. It also discusses on spine status in neurodegenerative diseases and the impact of nootropics and neuroprotective agents on the functional restoration of dendritic spines.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India.
| | - A G Rathipriya
- Food and Brain Research Foundation, Chennai, Tamil Nadu, India
| | - Srinivasa Rao Bolla
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Damam, Saudi Arabia
| | - Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Arehally Marappa Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Thamilarasan Manivasagam
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | - Arokiasamy Justin Thenmozhi
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
| | - Gilles J Guillemin
- Neuropharmacology Group, Faculty of Medicine and Health Sciences, Deb Bailey MND Research Laboratory, Macquarie University, Sydney, NSW 2109, Australia
| | - Ramesh Chandra
- Department of Chemistry, Ambedkar Centre for BioMedical Research, Delhi University, Delhi 110007, India
| | - Meena Kishore Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, 107, Wiggins Road, Saskatoon, SK S7N 5C9, Canada.
| |
Collapse
|
28
|
Chamma I, Sainlos M, Thoumine O. Biophysical mechanisms underlying the membrane trafficking of synaptic adhesion molecules. Neuropharmacology 2019; 169:107555. [PMID: 30831159 DOI: 10.1016/j.neuropharm.2019.02.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 01/13/2023]
Abstract
Adhesion proteins play crucial roles at synapses, not only by providing a physical trans-synaptic linkage between axonal and dendritic membranes, but also by connecting to functional elements including the pre-synaptic neurotransmitter release machinery and post-synaptic receptors. To mediate these functions, adhesion proteins must be organized on the neuronal surface in a precise and controlled manner. Recent studies have started to describe the mobility, nanoscale organization, and turnover rate of key synaptic adhesion molecules including cadherins, neurexins, neuroligins, SynCAMs, and LRRTMs, and show that some of these proteins are highly mobile in the plasma membrane while others are confined at sub-synaptic compartments, providing evidence for different regulatory pathways. In this review article, we provide a biophysical view of the diffusional trapping of adhesion molecules at synapses, involving both extracellular and intracellular protein interactions. We review the methodology underlying these measurements, including biomimetic systems with purified adhesion proteins, means to perturb protein expression or function, single molecule imaging in cultured neurons, and analytical models to interpret the data. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Ingrid Chamma
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France
| | - Matthieu Sainlos
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France
| | - Olivier Thoumine
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
29
|
Donato A, Kagias K, Zhang Y, Hilliard MA. Neuronal sub-compartmentalization: a strategy to optimize neuronal function. Biol Rev Camb Philos Soc 2019; 94:1023-1037. [PMID: 30609235 PMCID: PMC6617802 DOI: 10.1111/brv.12487] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/14/2022]
Abstract
Neurons are highly polarized cells that consist of three main structural and functional domains: a cell body or soma, an axon, and dendrites. These domains contain smaller compartments with essential roles for proper neuronal function, such as the axonal presynaptic boutons and the dendritic postsynaptic spines. The structure and function of these compartments have now been characterized in great detail. Intriguingly, however, in the last decade additional levels of compartmentalization within the axon and the dendrites have been identified, revealing that these structures are much more complex than previously thought. Herein we examine several types of structural and functional sub-compartmentalization found in neurons of both vertebrates and invertebrates. For example, in mammalian neurons the axonal initial segment functions as a sub-compartment to initiate the action potential, to select molecules passing into the axon, and to maintain neuronal polarization. Moreover, work in Drosophila melanogaster has shown that two distinct axonal guidance receptors are precisely clustered in adjacent segments of the commissural axons both in vivo and in vitro, suggesting a cell-intrinsic mechanism underlying the compartmentalized receptor localization. In Caenorhabditis elegans, a subset of interneurons exhibits calcium dynamics that are localized to specific sections of the axon and control the gait of navigation, demonstrating a regulatory role of compartmentalized neuronal activity in behaviour. These findings have led to a number of new questions, which are important for our understanding of neuronal development and function. How are these sub-compartments established and maintained? What molecular machinery and cellular events are involved? What is their functional significance for the neuron? Here, we reflect on these and other key questions that remain to be addressed in this expanding field of biology.
Collapse
Affiliation(s)
- Alessandra Donato
- Clem Jones Centre for Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Konstantinos Kagias
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, U.S.A
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, U.S.A
| | - Massimo A Hilliard
- Clem Jones Centre for Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
30
|
Lin AW, Gill KK, Castañeda MS, Matucci I, Eder N, Claxton S, Flynn H, Snijders AP, George R, Ultanir SK. Chemical genetic identification of GAK substrates reveals its role in regulating Na +/K +-ATPase. Life Sci Alliance 2018; 1:e201800118. [PMID: 30623173 PMCID: PMC6312924 DOI: 10.26508/lsa.201800118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/15/2022] Open
Abstract
Novel GAK phosphorylation targets are identified using chemical genetic methods. One of the substrates is the α subunit of the Na+/K+-ATPase, phosphorylation of which is necessary for its surface trafficking from endosomes. Conserved functions of NAK family kinases are described. Cyclin G–associated kinase (GAK) is a ubiquitous serine/threonine kinase that facilitates clathrin uncoating during vesicle trafficking. GAK phosphorylates a coat adaptor component, AP2M1, to help achieve this function. GAK is also implicated in Parkinson's disease through genome-wide association studies. However, GAK's role in mammalian neurons remains unclear, and insight may come from identification of further substrates. Employing a chemical genetics method, we show here that the sodium potassium pump (Na+/K+-ATPase) α-subunit Atp1a3 is a GAK target and that GAK regulates Na+/K+-ATPase trafficking to the plasma membrane. Whole-cell patch clamp recordings from CA1 pyramidal neurons in GAK conditional knockout mice show a larger change in resting membrane potential when exposed to the Na+/K+-ATPase blocker ouabain, indicating compromised Na+/K+-ATPase function in GAK knockouts. Our results suggest a modulatory role for GAK via phosphoregulation of substrates such as Atp1a3 during cargo trafficking.
Collapse
Affiliation(s)
- Amy W Lin
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Kalbinder K Gill
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | | | - Irene Matucci
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Noreen Eder
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom.,Mass Spectrometry Platform, The Francis Crick Institute, London, United Kingdom
| | - Suzanne Claxton
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Helen Flynn
- Mass Spectrometry Platform, The Francis Crick Institute, London, United Kingdom
| | | | - Roger George
- Protein Purification Facility, The Francis Crick Institute, London, United Kingdom
| | - Sila K Ultanir
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
31
|
Zhou F, Xie J, Zhang S, Yin G, Gao Y, Zhang Y, Bo D, Li Z, Liu S, Feng C, Fan G. Lead, cadmium, arsenic, and mercury combined exposure disrupted synaptic homeostasis through activating the Snk-SPAR pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 163:674-684. [PMID: 30099283 DOI: 10.1016/j.ecoenv.2018.07.116] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/19/2018] [Accepted: 07/28/2018] [Indexed: 06/08/2023]
Abstract
Lead (Pb), cadmium (Cd), arsenic (As), and mercury (Hg) are among the leading toxic agents detected in the environment, and they have also been detected simultaneously in blood, serum, and urine samples of the general population. Meanwhile early neurologic effects and multiple interactions of Pb, Cd, As, and Hg had been found in children from environmentally polluted area. However, the current studies of these four metals were mostly limited to the interactions between any two metals, whereas the interaction characteristics between any three and four metals were rarely studied. In our study, we firstly explored the characteristics of the neurotoxic interactions among these four elements in nerve cells with factorial designs. The results showed that Pb+Cd+As+Hg co-exposure had a synergistic neurotoxic effect that was more severe than that induced by any two or three metals, when their individual metals were at human environmental exposure (in the blood of U.S. population) relevant levels and below no observed adverse effect levels (NOAELs). Therefore, Pb+Cd+As+Hg co-exposure at human environmental exposure relevant levels were further selected to examine synaptic homeostasis as the cellular and molecular foundation of learning and memory. We reported for the first time that Pb+Cd+As+Hg co-exposure induced dose-dependent decreases of the dendritic lengths and branching, as well as spine density and mature phenotype in primary hippocampal neurons, and the stimulated neurite outgrowths in NGF-differentiated PC12 cells. And the above synaptic homeostasis disruption was associated with serum induced kinase (Snk)-spine associated Rap GTPase activating protein (SPAR) pathway. Our study suggests that human environmental Pb, Cd, As, and Hg co-exposure has the potential to evoke synergistic neurotoxicity even if their individual metals are below NOAELs, which reinforces the need to control and regulate potential sources of metal contamination.
Collapse
Affiliation(s)
- Fankun Zhou
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Jie Xie
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Shuyun Zhang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Guangming Yin
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Yanyan Gao
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Yuanyuan Zhang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Dandan Bo
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Zongguang Li
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Sisi Liu
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Chang Feng
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Guangqin Fan
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
32
|
Kim S, Zhang Y, Jin C, Lee Y, Kim Y, Han K. Emerging roles of Lys63-linked polyubiquitination in neuronal excitatory postsynapses. Arch Pharm Res 2018; 42:285-292. [DOI: 10.1007/s12272-018-1081-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022]
|
33
|
Phosphorylation of synaptic GTPase-activating protein (synGAP) by polo-like kinase (Plk2) alters the ratio of its GAP activity toward HRas, Rap1 and Rap2 GTPases. Biochem Biophys Res Commun 2018; 503:1599-1604. [PMID: 30049443 DOI: 10.1016/j.bbrc.2018.07.087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/18/2018] [Indexed: 11/20/2022]
Abstract
SynGAP is a Ras and Rap GTPase-activating protein (GAP) found in high concentration in the postsynaptic density (PSD) fraction from mammalian forebrain where it binds to PDZ domains of PSD-95. Phosphorylation of pure recombinant synGAP by Ca2+/calmodulin-dependent protein kinase II (CaMKII) shifts the balance of synGAP's GAP activity toward inactivation of Rap1; whereas phosphorylation by cyclin-dependent kinase 5 (CDK5) has the opposite effect, shifting the balance toward inactivation of HRas. These shifts in balance contribute to regulation of the numbers of surface AMPA receptors, which rise during synaptic potentiation (CaMKII) and fall during synaptic scaling (CDK5). Polo-like kinase 2 (Plk2/SNK), like CDK5, contributes to synaptic scaling. These two kinases act in concert to reduce the number of surface AMPA receptors following elevated neuronal activity by tagging spine-associated RapGAP protein (SPAR) for degradation, thus raising the level of activated Rap. Here we show that Plk2 also phosphorylates and regulates synGAP. Phosphorylation of synGAP by Plk2 stimulates its GAP activity toward HRas by 65%, and toward Rap1 by 16%. Simultaneous phosphorylation of synGAP by Plk2 and CDK5 at distinct sites produces an additive increase in GAP activity toward HRas (∼230%) and a smaller, non-additive increase in activity toward Rap1 (∼15%). Dual phosphorylation also produces an increase in GAP activity toward Rap2 (∼40-50%), an effect not produced by either kinase alone. As we previously observed for CDK5, addition of Ca2+/CaM causes a substrate-directed doubling of the rate and stoichiometry of phosphorylation of synGAP by Plk2, targeting residues also phosphorylated by CaMKII. In summary, phosphorylation by Plk2, like CDK5, shifts the ratio of GAP activity of synGAP to produce a greater decrease in active Ras than in active Rap, which would produce a shift toward a decrease in the number of surface AMPA receptors in neuronal dendrites.
Collapse
|
34
|
Abstract
Mutations in the cereblon (CRBN) gene cause human intellectual disability, one of the most common cognitive disorders. However, the molecular mechanisms of CRBN-related intellectual disability remain poorly understood. We investigated the role of CRBN in synaptic function and animal behavior using male mouse and Drosophila models. Crbn knock-out (KO) mice showed normal brain and spine morphology as well as intact synaptic plasticity; however, they also exhibited decreases in synaptic transmission and presynaptic release probability exclusively in excitatory synapses. Presynaptic function was impaired not only by loss of CRBN expression, but also by expression of pathogenic CRBN mutants (human R419X mutant and Drosophila G552X mutant). We found that the BK channel blockers paxilline and iberiotoxin reversed this decrease in presynaptic release probability in Crbn KO mice. In addition, paxilline treatment also restored normal cognitive behavior in Crbn KO mice. These results strongly suggest that increased BK channel activity is the pathological mechanism of intellectual disability in CRBN mutations.SIGNIFICANCE STATEMENTCereblon (CRBN), a well known target of the immunomodulatory drug thalidomide, was originally identified as a gene that causes human intellectual disability when mutated. However, the molecular mechanisms of CRBN-related intellectual disability remain poorly understood. Based on the idea that synaptic abnormalities are the most common factor in cognitive dysfunction, we monitored the synaptic structure and function of Crbn knock-out (KO) animals to identify the molecular mechanisms of intellectual disability. Here, we found that Crbn KO animals showed cognitive deficits caused by enhanced BK channel activity and reduced presynaptic glutamate release. Our findings suggest a physiological pathomechanism of the intellectual disability-related gene CRBN and will contribute to the development of therapeutic strategies for CRBN-related intellectual disability.
Collapse
|
35
|
The Adiponectin Homolog Osmotin Enhances Neurite Outgrowth and Synaptic Complexity via AdipoR1/NgR1 Signaling in Alzheimer’s Disease. Mol Neurobiol 2018; 55:6673-6686. [DOI: 10.1007/s12035-017-0847-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/19/2017] [Indexed: 01/05/2023]
|
36
|
Dutta SM, Hadley MM, Peterman S, Jewell JS, Duncan VD, Britten RA. Quantitative Proteomic Analysis of the Hippocampus of Rats with GCR-Induced Spatial Memory Impairment. Radiat Res 2017; 189:136-145. [PMID: 29206597 DOI: 10.1667/rr14822.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
NASA is planning future missions to Mars, which will result in astronauts being exposed to ∼13 cGy/year of galactic cosmic radiation (GCR). Previous ground-based experiments have demonstrated that low (15 cGy) doses of 1 GeV/n 56Fe ions impair hippocampus-dependent spatial memory in rats. However, some irradiated rats maintain a spatial memory performance comparable to that seen in the sham-irradiated rats, suggesting that some of these animals are able to ameliorate the deleterious effects of the GCR, while others are not. This rat model provides a unique opportunity to increase our understanding of how GCR affects neurophysiology, what adaptive responses can be invoked to prevent the emergence of GCR-induced spatial memory impairment, as well as the pathways that are altered when spatial memory impairment occurs. A label-free, unbiased proteomic profiling approach involving quantitative protein/peptide profiling followed by Cytoscape analysis has established the composition of the hippocampal proteome in male Wistar rats after exposure to 15 cGy of 1 GeV/n 56Fe, and identified proteins whose expression is altered with respect to: 1. radiation exposure and 2. impaired spatial memory performance. We identified 30 proteins that were classified as "GCR exposure marker" (GEM) proteins (expressed solely or at higher levels in the irradiated rats but not related to spatial memory performance), most notably CD98, Cadps and GMFB. Conversely, there were 252 proteins that were detected only in the sham-irradiated samples, i.e., they were not detected in either of the irradiated cohorts; of these 10% have well-documented roles in neurotransmission. The second aspect of our data mining was to identify proteins whose expression was associated with either impaired or functional spatial memory. While there are multiple changes in the hippocampal proteome in the irradiated rats that have impaired spatial memory performance, with 203 proteins being detected (or upregulated) only in these rats, it would appear that spatial memory impairment may also arise from an inability of these rats to express "good spatial memory" (GSM) proteins, many of which play an important role in neuronal homeostasis and function, axonogenesis, presynaptic membrane organization and G-protein coupled receptor (GCPR) signaling. It may be possible to use this knowledge to develop two alternative countermeasure strategies, one that preserves critical pathways prophylactically and one that invokes restorative pathways after GCR exposure.
Collapse
Affiliation(s)
- Sucharita M Dutta
- a Leroy T. Canoles Jr. Cancer Research Center and.,b Departments of Microbiology and Molecular Cell Biology and
| | - Melissa M Hadley
- c Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507; and
| | - Scott Peterman
- d BRIMS, Thermo Fisher Scientific, Cambridge, Massachusetts 02139
| | - Jessica S Jewell
- c Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507; and
| | - Vania D Duncan
- c Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507; and
| | - Richard A Britten
- a Leroy T. Canoles Jr. Cancer Research Center and.,b Departments of Microbiology and Molecular Cell Biology and.,c Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507; and
| |
Collapse
|
37
|
Avila J, Llorens-Martín M, Pallas-Bazarra N, Bolós M, Perea JR, Rodríguez-Matellán A, Hernández F. Cognitive Decline in Neuronal Aging and Alzheimer's Disease: Role of NMDA Receptors and Associated Proteins. Front Neurosci 2017; 11:626. [PMID: 29176942 PMCID: PMC5687061 DOI: 10.3389/fnins.2017.00626] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/26/2017] [Indexed: 01/01/2023] Open
Abstract
Molecular changes associated with neuronal aging lead to a decrease in cognitive capacity. Here we discuss these alterations at the level of brain regions, brain cells, and brain membrane and cytoskeletal proteins with an special focus in NMDA molecular changes through aging and its effect in cognitive decline and Alzheimer disease. Here, we propose that some neurodegenerative disorders, like Alzheimer's disease (AD), are characterized by an increase and acceleration of some of these changes.
Collapse
Affiliation(s)
- Jesús Avila
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autonoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Llorens-Martín
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autonoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Noemí Pallas-Bazarra
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autonoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Marta Bolós
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autonoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Juan R Perea
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autonoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Alberto Rodríguez-Matellán
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autonoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autonoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
38
|
mTORC and ProSAPiP1: How Alcohol Changes Synapses of Reward Circuitry. Neuron 2017; 96:6-8. [PMID: 28957677 DOI: 10.1016/j.neuron.2017.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alcohol addiction is characterized by broad and persistent changes in brain function, but the underlying neural adaptations remain largely unknown. In this issue of Neuron, Laguesse et al. (2017) describe a neural mechanism through which long-term alcohol exposure induces structural and synaptic adaptations that promote excessive alcohol use.
Collapse
|
39
|
Laguesse S, Morisot N, Shin JH, Liu F, Adrover MF, Sakhai SA, Lopez MF, Phamluong K, Griffin WC, Becker HC, Bender KJ, Alvarez VA, Ron D. Prosapip1-Dependent Synaptic Adaptations in the Nucleus Accumbens Drive Alcohol Intake, Seeking, and Reward. Neuron 2017; 96:145-159.e8. [PMID: 28890345 PMCID: PMC6014831 DOI: 10.1016/j.neuron.2017.08.037] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/13/2017] [Accepted: 08/24/2017] [Indexed: 12/15/2022]
Abstract
The mammalian target of rapamycin complex 1 (mTORC1), a transducer of local dendritic translation, participates in learning and memory processes as well as in mechanisms underlying alcohol-drinking behaviors. Using an unbiased RNA-seq approach, we identified Prosapip1 as a novel downstream target of mTORC1 whose translation and consequent synaptic protein expression are increased in the nucleus accumbens (NAc) of mice excessively consuming alcohol. We demonstrate that alcohol-dependent increases in Prosapip1 levels promote the formation of actin filaments, leading to changes in dendritic spine morphology of NAc medium spiny neurons (MSNs). We further demonstrate that Prosapip1 is required for alcohol-dependent synaptic localization of GluA2 lacking AMPA receptors in NAc shell MSNs. Finally, we present data implicating Prosapip1 in mechanisms underlying alcohol self-administration and reward. Together, these data suggest that Prosapip1 in the NAc is a molecular transducer of structural and synaptic alterations that drive and/or maintain excessive alcohol use.
Collapse
Affiliation(s)
- Sophie Laguesse
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Nadege Morisot
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Jung Hoon Shin
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute of Alcohol Abuse and Alcoholism, US National Institutes of Health, Bethesda, MD, USA
| | - Feng Liu
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Martin F Adrover
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute of Alcohol Abuse and Alcoholism, US National Institutes of Health, Bethesda, MD, USA
| | - Samuel A Sakhai
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California, San Francisco, CA, USA
| | - William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA; Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA; RHJ Department of Veterans Affairs Medical Center, Charleston, SC, USA
| | - Kevin J Bender
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute of Alcohol Abuse and Alcoholism, US National Institutes of Health, Bethesda, MD, USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, CA, USA.
| |
Collapse
|
40
|
Proteasome-independent polyubiquitin linkage regulates synapse scaffolding, efficacy, and plasticity. Proc Natl Acad Sci U S A 2017; 114:E8760-E8769. [PMID: 28973854 DOI: 10.1073/pnas.1620153114] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ubiquitination-directed proteasomal degradation of synaptic proteins, presumably mediated by lysine 48 (K48) of ubiquitin, is a key mechanism in synapse and neural circuit remodeling. However, more than half of polyubiquitin (polyUb) species in the mammalian brain are estimated to be non-K48; among them, the most abundant is Lys 63 (K63)-linked polyUb chains that do not tag substrates for degradation but rather modify their properties and activity. Virtually nothing is known about the role of these nonproteolytic polyUb chains at the synapse. Here we report that K63-polyUb chains play a significant role in postsynaptic protein scaffolding and synaptic strength and plasticity. We found that the postsynaptic scaffold PSD-95 (postsynaptic density protein 95) undergoes K63 polyubiquitination, which markedly modifies PSD-95's scaffolding potentials, enables its synaptic targeting, and promotes synapse maturation and efficacy. TNF receptor-associated factor 6 (TRAF6) is identified as a direct E3 ligase for PSD-95, which, together with the E2 complex Ubc13/Uev1a, assembles K63-chains on PSD-95. In contrast, CYLD (cylindromatosis tumor-suppressor protein), a K63-specific deubiquitinase enriched in postsynaptic densities, cleaves K63-chains from PSD-95. We found that neuronal activity exerts potent control of global and synaptic K63-polyUb levels and, through NMDA receptors, drives rapid, CYLD-mediated PSD-95 deubiquitination, mobilizing and depleting PSD-95 from synapses. Silencing CYLD in hippocampal neurons abolishes NMDA-induced chemical long-term depression. Our results unveil a previously unsuspected role for nonproteolytic polyUb chains in the synapse and illustrate a mechanism by which a PSD-associated K63-linkage-specific ubiquitin machinery acts on a major postsynaptic scaffold to regulate synapse organization, function, and plasticity.
Collapse
|
41
|
Wu Q, Sun M, Bernard LP, Zhang H. Postsynaptic density 95 (PSD-95) serine 561 phosphorylation regulates a conformational switch and bidirectional dendritic spine structural plasticity. J Biol Chem 2017; 292:16150-16160. [PMID: 28790172 DOI: 10.1074/jbc.m117.782490] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/28/2017] [Indexed: 01/12/2023] Open
Abstract
Postsynaptic density 95 (PSD-95) is a major synaptic scaffolding protein that plays a key role in bidirectional synaptic plasticity, which is a process important for learning and memory. It is known that PSD-95 shows increased dynamics upon induction of plasticity. However, the underlying structural and functional changes in PSD-95 that mediate its role in plasticity remain unclear. Here we show that phosphorylation of PSD-95 at Ser-561 in its guanylate kinase (GK) domain, which is mediated by the partitioning-defective 1 (Par1) kinases, regulates a conformational switch and is important for bidirectional plasticity. Using a fluorescence resonance energy transfer (FRET) biosensor, we show that a phosphomimetic mutation of Ser-561 promotes an intramolecular interaction between GK and the nearby Src homology 3 (SH3) domain, leading to a closed conformation, whereas a non-phosphorylatable S561A mutation or inhibition of Par1 kinase activity decreases SH3-GK interaction, causing PSD-95 to adopt an open conformation. In addition, S561A mutation facilitates the interaction between PSD-95 and its binding partners. Fluorescence recovery after photobleaching imaging reveals that the S561A mutant shows increased stability, whereas the phosphomimetic S561D mutation increases PSD-95 dynamics at the synapse. Moreover, molecular replacement of endogenous PSD-95 with the S561A mutant blocks dendritic spine structural plasticity during chemical long-term potentiation and long-term depression. Endogenous Ser-561 phosphorylation is induced by synaptic NMDA receptor activation, and the SH3-GK domains exhibit a Ser-561 phosphorylation-dependent switch to a closed conformation during synaptic plasticity. Our results provide novel mechanistic insight into the regulation of PSD-95 in dendritic spine structural plasticity through phosphorylation-mediated regulation of protein dynamics and conformation.
Collapse
Affiliation(s)
- Qian Wu
- From the Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
| | - Miao Sun
- From the Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
| | - Laura P Bernard
- From the Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
| | - Huaye Zhang
- From the Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
| |
Collapse
|
42
|
Shah B, Püschel AW. Regulation of Rap GTPases in mammalian neurons. Biol Chem 2017; 397:1055-69. [PMID: 27186679 DOI: 10.1515/hsz-2016-0165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022]
Abstract
Small GTPases are central regulators of many cellular processes. The highly conserved Rap GTPases perform essential functions in the mammalian nervous system during development and in mature neurons. During neocortical development, Rap1 is required to regulate cadherin- and integrin-mediated adhesion. In the adult nervous system Rap1 and Rap2 regulate the maturation and plasticity of dendritic spine and synapses. Although genetic studies have revealed important roles of Rap GTPases in neurons, their regulation by guanine nucleotide exchange factors (GEFs) that activate them and GTPase activating proteins (GAPs) that inactivate them by stimulating their intrinsic GTPase activity is just beginning to be explored in vivo. Here we review how GEFs and GAPs regulate Rap GTPases in the nervous system with a focus on their in vivo function.
Collapse
|
43
|
Sui H, Zhan L, Niu X, Liang L, Li X. The SNK and SPAR signaling pathway changes in hippocampal neurons treated with amyloid-beta peptide in vitro. Neuropeptides 2017; 63:43-48. [PMID: 28400058 DOI: 10.1016/j.npep.2017.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 02/19/2017] [Accepted: 03/16/2017] [Indexed: 11/24/2022]
Abstract
Amyloid-β peptide (Aβ) is believed to be a primary cause of Alzheimer's disease. Many studies have demonstrated that Aβ causes morphological and functional alterations of dendritic spines, leading to synaptic dysfunction, but the effect of Aβ on damage to synaptic functions is not fully understood. Spine-associated Rap guanosine triphosphatase-activating protein (SPAR) is an important regulator of activity-dependent remodeling of synapses and is critically involved in both mature dendritic spine formation and the maintenance of spine maturity. Serum-inducible kinase (SNK) is an activity-inducible member of the polo-like family of serine/threonine kinases. Coordinated regulation of Ras and Rap by SNK is critical for homeostatic plasticity and memory. A previous study in which rats were injected with Aβ1-40 into the hippocampus showed that the SNK and SPAR signaling pathway may play a crucial role in Aβ-induced excitotoxic damage in the central nervous system by regulating synaptic stability. The present study was designed to investigate whether the SNK and SPAR signaling pathway was involved in Aβ-induced neurotoxicity in rat primary neurons. We measured mRNA and protein expression levels of SNK and SPAR in primary hippocampal neurons following Aβ treatment and used RNA interference to knockdown SNK to investigate the underlying mechanism. Expression of SNK and SPAR was altered by Aβ treatment, indicating that the SNK and SPAR signaling pathways may be involved in the damage to dendritic spines in hippocampal neurons induced by Aβ.
Collapse
Affiliation(s)
- Hua Sui
- Institute of Basic Research of Integrative Medicine, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Libin Zhan
- The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, Liaoning, China; College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China.
| | - Xinping Niu
- Department of Cardiology, Dalian Second People's Hospital, Dalian 116011, Liaoning, China
| | - Lina Liang
- Institute of Basic Research of Integrative Medicine, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Xin Li
- Institute of Basic Research of Integrative Medicine, Dalian Medical University, Dalian 116044, Liaoning, China
| |
Collapse
|
44
|
Shah K, Rossie S. Tale of the Good and the Bad Cdk5: Remodeling of the Actin Cytoskeleton in the Brain. Mol Neurobiol 2017; 55:3426-3438. [PMID: 28502042 DOI: 10.1007/s12035-017-0525-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/06/2017] [Indexed: 11/24/2022]
Abstract
Cdk5 kinase, a cyclin-dependent kinase family member, is a key regulator of cytoskeletal remodeling in the brain. Cdk5 is essential for brain development during embryogenesis. After birth, it is essential for numerous neuronal processes such as learning and memory formation, drug addiction, pain signaling, and long-term behavior changes, all of which rely on rapid alterations in the cytoskeleton. Cdk5 activity is deregulated in various brain disorders including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and ischemic stroke, resulting in profound remodeling of the neuronal cytoskeleton, loss of synapses, and ultimately neurodegeneration. This review focuses on the "good and bad" Cdk5 in the brain and its pleiotropic contribution in regulating neuronal actin cytoskeletal remodeling. A vast majority of physiological and pathological Cdk5 substrates are associated with the actin cytoskeleton. Thus, our special emphasis is on the numerous Cdk5 substrates identified in the past two decades such as ephexin1, p27, Mst3, CaMKv, kalirin-7, RasGRF2, Pak1, WAVE1, neurabin-1, TrkB, 5-HT6R, talin, drebrin, synapsin I, synapsin III, CRMP1, GKAP, SPAR, PSD-95, and LRRK2. These substrates have unraveled the molecular mechanisms by which Cdk5 plays divergent roles in regulating neuronal actin cytoskeletal dynamics both in healthy and diseased states.
Collapse
Affiliation(s)
- Kavita Shah
- Department of Chemistry and Purdue University Center of Cancer Research, Purdue University, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| | - Sandra Rossie
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
45
|
ZBP1 phosphorylation at serine 181 regulates its dendritic transport and the development of dendritic trees of hippocampal neurons. Sci Rep 2017; 7:1876. [PMID: 28500298 PMCID: PMC5431813 DOI: 10.1038/s41598-017-01963-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 04/07/2017] [Indexed: 11/23/2022] Open
Abstract
Local protein synthesis occurs in axons and dendrites of neurons, enabling fast and spatially restricted responses to a dynamically changing extracellular environment. Prior to local translation, mRNA that is to be translated is packed into ribonucleoprotein particles (RNPs) where RNA binding proteins ensure mRNA silencing and provide a link to molecular motors. ZBP1 is a component of RNP transport particles and is known for its role in the local translation of β-actin mRNA. Its binding to mRNA is regulated by tyrosine 396 phosphorylation, and this particular modification was shown to be vital for axonal growth and dendritic branching. Recently, additional phosphorylation of ZBP1 at serine 181 (Ser181) was described in non-neuronal cells. In the present study, we found that ZBP1 is also phosphorylated at Ser181 in neurons in a mammalian/mechanistic target of rapamycin complex 2-, Src kinase-, and mRNA binding-dependent manner. Furthermore, Ser181 ZBP1 phosphorylation was essential for the proper dendritic branching of hippocampal neurons that were cultured in vitro and for the proper ZBP1 dendritic distribution and motility.
Collapse
|
46
|
Wang PJ, Lin ST, Liu SH, Kuo KT, Hsu CH, Knepper MA, Yu MJ. Vasopressin-induced serine 269 phosphorylation reduces Sipa1l1 (signal-induced proliferation-associated 1 like 1)-mediated aquaporin-2 endocytosis. J Biol Chem 2017; 292:7984-7993. [PMID: 28336531 DOI: 10.1074/jbc.m117.779611] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/14/2017] [Indexed: 11/06/2022] Open
Abstract
The abundance of integral membrane proteins in the plasma membrane is determined by a dynamic balance between exocytosis and endocytosis, which can often be regulated by physiological stimuli. Here, we describe a mechanism that accounts for the ability of the peptide hormone vasopressin to regulate water excretion via a phosphorylation-dependent modulation of the PDZ domain-ligand interaction involving the water channel protein aquaporin-2. We discovered that the PDZ domain-containing protein Sipa1l1 (signal-induced proliferation-associated 1 like 1) binds to the cytoplasmic PDZ-ligand motif of aquaporin-2 and accelerates its endocytosis in the absence of vasopressin. Vasopressin-induced aquaporin-2 phosphorylation within the type I PDZ-ligand motif disrupted the interaction, in association with reduced aquaporin-2 endocytosis and prolonged plasma membrane aquaporin-2 retention. This phosphorylation-dependent alteration in the PDZ domain-ligand interaction was explained by 3D structural models, which showed a hormone-regulated mechanism that controls osmotic water transport and systemic water balance in mammals.
Collapse
Affiliation(s)
- Po-Jen Wang
- From the Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - Shu-Ting Lin
- From the Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - Shao-Hsuan Liu
- From the Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - Kuang-Ting Kuo
- From the Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - Chun-Hua Hsu
- the Department of Agricultural Chemistry, National Taiwan University, Taipei 10617, Taiwan, and
| | - Mark A Knepper
- the Systems Biology Center, NHLBI, National Institutes of Health, Bethesda, Maryland 20892-1603
| | - Ming-Jiun Yu
- From the Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei 10051, Taiwan,
| |
Collapse
|
47
|
Song JM, Sung YM, Nam JH, Yoon H, Chung A, Moffat E, Jung M, Pak DTS, Kim J, Hoe HS. A Mercaptoacetamide-Based Class II Histone Deacetylase Inhibitor Increases Dendritic Spine Density via RasGRF1/ERK Pathway. J Alzheimers Dis 2016; 51:591-604. [PMID: 26890742 DOI: 10.3233/jad-150717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The accumulation of amyloid-β (Aβ) leads to the loss of dendritic spines and synapses, which is hypothesized to cause cognitive impairments in Alzheimer's disease (AD) patients. In our previous study, we demonstrated that a novel mercaptoacetamide-based class II histone deacetylase inhibitor (HDACI), known as W2, decreased Aβ levels and improved learning and memory in mice. However, the underlying mechanism of this effect is unknown. OBJECTIVE Because dendritic spine formation is associated with cognitive performance, here we investigated whether HDACI W2 regulates dendritic spine density and its molecular mechanism of action. METHODS To examine the effect of HDACI W2 on dendritic spine density, we conducted morphological analysis of dendritic spines using GFP transfection and Golgi staining. In addition, to determine the molecular mechanism of W2 effects on spines, we measured the levels of mRNAs and proteins involved in the Ras signaling pathway using quantitative real-time PCR, immunocytochemistry, and western analysis. RESULTS We found that HDACI W2 altered dendritic spine density and morphology in vitro and in vivo. Additionally, W2 increased the mRNA or protein levels of Ras GRF1 and phospho-ERK. Moreover, knockdown of RasGRF1 and inhibition of ERK activity prevented the W2-mediated spinogenesis in primary hippocampal neurons. CONCLUSION Our Class II-selective HDACI W2 promotes the formation and growth of dendritic spines in a RasGRF1 and ERK dependent manner in primary hippocampal neurons.
Collapse
Affiliation(s)
- Jung Min Song
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - You Me Sung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Jin Han Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, Korea
| | - Hyejin Yoon
- Department of Neuroscience, Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Andrew Chung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Emily Moffat
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Mira Jung
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, USA
| | - Jungsu Kim
- Department of Neuroscience, Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.,Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, Korea
| |
Collapse
|
48
|
The TRPM7 interactome defines a cytoskeletal complex linked to neuroblastoma progression. Eur J Cell Biol 2016; 95:465-474. [DOI: 10.1016/j.ejcb.2016.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/16/2016] [Accepted: 06/23/2016] [Indexed: 01/27/2023] Open
|
49
|
Actin-Dependent Alterations of Dendritic Spine Morphology in Shankopathies. Neural Plast 2016; 2016:8051861. [PMID: 27795858 PMCID: PMC5067329 DOI: 10.1155/2016/8051861] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/30/2016] [Indexed: 12/11/2022] Open
Abstract
Shank proteins (Shank1, Shank2, and Shank3) act as scaffolding molecules in the postsynaptic density of many excitatory neurons. Mutations in SHANK genes, in particular SHANK2 and SHANK3, lead to autism spectrum disorders (ASD) in both human and mouse models. Shank3 proteins are made of several domains-the Shank/ProSAP N-terminal (SPN) domain, ankyrin repeats, SH3 domain, PDZ domain, a proline-rich region, and the sterile alpha motif (SAM) domain. Via various binding partners of these domains, Shank3 is able to bind and interact with a wide range of proteins including modulators of small GTPases such as RICH2, a RhoGAP protein, and βPIX, a RhoGEF protein for Rac1 and Cdc42, actin binding proteins and actin modulators. Dysregulation of all isoforms of Shank proteins, but especially Shank3, leads to alterations in spine morphogenesis, shape, and activity of the synapse via altering actin dynamics. Therefore, here, we highlight the role of Shank proteins as modulators of small GTPases and, ultimately, actin dynamics, as found in multiple in vitro and in vivo models. The failure to mediate this regulatory role might present a shared mechanism in the pathophysiology of autism-associated mutations, which leads to dysregulation of spine morphogenesis and synaptic signaling.
Collapse
|
50
|
Putative Cell Adhesion Membrane Protein Vstm5 Regulates Neuronal Morphology and Migration in the Central Nervous System. J Neurosci 2016; 36:10181-97. [PMID: 27683913 DOI: 10.1523/jneurosci.0541-16.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/16/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED During brain development, dynamic changes in neuronal membranes perform critical roles in neuronal morphogenesis and migration to create functional neural circuits. Among the proteins that induce membrane dynamics, cell adhesion molecules are important in neuronal membrane plasticity. Here, we report that V-set and transmembrane domain-containing protein 5 (Vstm5), a cell-adhesion-like molecule belonging to the Ig superfamily, was found in mouse brain. Knock-down of Vstm5 in cultured hippocampal neurons markedly reduced the complexity of dendritic structures, as well as the number of dendritic filopodia. Vstm5 also regulates neuronal morphology by promoting dendritic protrusions that later develop into dendritic spines. Using electroporation in utero, we found that Vstm5 overexpression delayed neuronal migration and induced multiple branches in leading processes during corticogenesis. These results indicate that Vstm5 is a new cell-adhesion-like molecule and is critically involved in synaptogenesis and corticogenesis by promoting neuronal membrane dynamics. SIGNIFICANCE STATEMENT Neuronal migration and morphogenesis play critical roles in brain development and function. In this study, we demonstrate for the first time that V-set and transmembrane domain-containing protein 5 (Vstm5), a putative cell adhesion membrane protein, modulates both the position and complexity of central neurons by altering their membrane morphology and dynamics. Vstm5 is also one of the target genes responsible for variations in patient responses to treatments for major depressive disorder. Our results provide the first evidence that Vstm5 is a novel factor involved in the modulation of the neuronal membrane and a critical element in normal neural circuit formation during mammalian brain development.
Collapse
|