1
|
Matsumoto N. Endogenously generated patterns of neural activity sculpt axon connectivity. Neurosci Res 2025:S0168-0102(25)00085-9. [PMID: 40389064 DOI: 10.1016/j.neures.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/21/2025]
Abstract
Neural activity is crucial in establishing functional circuit connectivity in the central nervous system. Prior to the onset of sensory experience, sensory organs spontaneously generate patterned neural activity, which is essential for sculpting and refining immature circuit connectivity coordinating functional and physiological responses to the external world in advance. How these endogenous patterns of neural activity drive circuit refinement is a major long-standing question; however, it has been impeded, at least partly, by technical difficulties in visualizing circuit refinement and patterned spontaneous activity in living animals. In this review, I discuss recent progress in visualizing circuit refinement processes and patterned spontaneous activity at the single-axon level in the mammalian visual system and consider how endogenous patterns of spontaneous activity drive fine-scale axon refinement during development.
Collapse
Affiliation(s)
- Naoyuki Matsumoto
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics (NIG), Mishima, Shizuoka 411-8540, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka 411-8540, Japan.
| |
Collapse
|
2
|
Qneibi M, Bdir S, Bdair M, Aldwaik SA, Heeh M, Sandouka D, Idais T. Exploring the role of AMPA receptor auxiliary proteins in synaptic functions and diseases. FEBS J 2025; 292:2433-2478. [PMID: 39394632 DOI: 10.1111/febs.17287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/21/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) ionotropic glutamate receptors (AMPARs) mediate rapid excitatory synaptic transmission in the mammalian brain, primarily driven by the neurotransmitter glutamate. The modulation of AMPAR activity, particularly calcium-permeable AMPARs (CP-AMPARs), is crucially influenced by various auxiliary subunits. These subunits are integral membrane proteins that bind to the receptor's core and modify its functional properties, including ion channel kinetics and receptor trafficking. This review comprehensively catalogs all known AMPAR auxiliary proteins, providing vital insights into the biochemical mechanisms governing synaptic modulation and the specific impact of CP-AMPARs compared to their calcium-impermeable AMPA receptor (CI-AMPARs). Understanding the complex interplay between AMPARs and their auxiliary subunits in different brain regions is essential for elucidating their roles in cognitive functions such as learning and memory. Importantly, alterations in these auxiliary proteins' expression, function or interactions have been implicated in various neurological disorders. Aberrant signaling through CP-AMPARs, in particular, is associated with severe synaptic dysfunctions across neurodevelopmental, neurodegenerative and psychiatric conditions. Targeting the distinct properties of AMPAR-auxiliary subunit complexes, especially those involving CP-AMPARs, could disclose new therapeutic strategies, potentially allowing for more precise interventions in treating complex neuronal disorders.
Collapse
Affiliation(s)
- Mohammad Qneibi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Sosana Bdir
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammad Bdair
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Samia Ammar Aldwaik
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | | | - Dana Sandouka
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Tala Idais
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| |
Collapse
|
3
|
Almodóvar-Payá C, París-Gómez I, Latorre-Guardia M, Guardiola-Ripoll M, Catalán R, Arias B, Penadés R, Fatjó-Vilas M. NRN1 genetic variability and methylation changes as biomarkers for cognitive remediation therapy response in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111175. [PMID: 39426559 DOI: 10.1016/j.pnpbp.2024.111175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/20/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Cognitive remediation therapy (CRT) demonstrates potential in enhancing cognitive function in schizophrenia (SZ), though the identification of molecular biomarkers remains challenging. The Neuritin-1 gene (NRN1) emerges as a promising candidate gene due to its association with SZ, cognitive performance and response to neurotherapeutic treatments. We aimed to investigate whether NRN1 genetic variability and methylation changes following CRT are related to cognitive improvements. Twenty-five SZ patients were randomly assigned to CRT or treatment-as-usual (TAU) groups, with cognitive function and NRN1 methylation assessed pre- and post-intervention using the MATRICS Consensus Cognitive Battery and EpiTYPER. Besides, eleven NRN1 polymorphisms were genotyped. Methylation changes (Δm = post - pre) were analyzed via sparse Partial Least Square Discriminant Analysis (sPLS-DA) to identify latent components (LCs) distinguishing CRT from TAU. To further explore methylation patterns of these LCs, CpG units were grouped into two subsets, yielding Δm means for those with increased and decreased methylation. Cognitive changes (Δcog = post - pre) were used to identify CRT improvers (CRT-I, Δcog ≥ 1), and the association between methylation changes and cognitive improvements post-therapy was also tested. We identified two LCs that differentiated CRT from TAU with a classification error rate of 0.28. The main component, LC1, included 25 CpG units. The subsets of CpG units with increased and decreased post-therapy methylation differed significantly between the two treatment arms, suggesting that differences were not merely data-driven but reflected meaningful biological variation. Additionally, CpG units linked to therapy were also associated with cognitive improvement, with LC1 and the subset of CpG units showing increased methylation post-therapy distinguishing CRT-I from the rest of the patients across multiple cognitive domains. Furthermore, the effect of LC1 on speed processing improvement after CRT was enhanced by considering the NRN1-rs9405890 polymorphism. Notably, these CpG units, particularly those with increased methylation after CRT, overlapped with key gene regulatory elements. Our model, integrating genetics and epigenetics, boosts the understanding of CRT response variability and highlights this multi-level approach as a promising strategy for identifying potential NRN1-related biomarkers of CRT effects, though further studies with larger samples are needed.
Collapse
Affiliation(s)
- Carmen Almodóvar-Payá
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | | | - Mariona Latorre-Guardia
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | | | - Rosa Catalán
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Departament de Medicina, Campus Clínic, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; Barcelona Clinic Schizophrenia Unit (BCSU), Hospital Clínic, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Bárbara Arias
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Rafael Penadés
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Barcelona Clinic Schizophrenia Unit (BCSU), Hospital Clínic, Barcelona, Spain; Departament de Psicologia Clínica i Psicobiologia, Facultat de Psicologia, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Mar Fatjó-Vilas
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Yu H, Nishio H, Barbi J, Mitchell-Flack M, Vignali PDA, Zheng Y, Lebid A, Chang KY, Fu J, Higgins M, Huang CT, Zhang X, Li Z, Blosser L, Tam A, Drake C, Pardoll D. Neurotrophic factor Neuritin modulates T cell electrical and metabolic state for the balance of tolerance and immunity. eLife 2024; 13:RP96812. [PMID: 39565188 DOI: 10.7554/elife.96812] [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] [Indexed: 11/21/2024] Open
Abstract
The adaptive T cell response is accompanied by continuous rewiring of the T cell's electric and metabolic state. Ion channels and nutrient transporters integrate bioelectric and biochemical signals from the environment, setting cellular electric and metabolic states. Divergent electric and metabolic states contribute to T cell immunity or tolerance. Here, we report in mice that neuritin (Nrn1) contributes to tolerance development by modulating regulatory and effector T cell function. Nrn1 expression in regulatory T cells promotes its expansion and suppression function, while expression in the T effector cell dampens its inflammatory response. Nrn1 deficiency in mice causes dysregulation of ion channel and nutrient transporter expression in Treg and effector T cells, resulting in divergent metabolic outcomes and impacting autoimmune disease progression and recovery. These findings identify a novel immune function of the neurotrophic factor Nrn1 in regulating the T cell metabolic state in a cell context-dependent manner and modulating the outcome of an immune response.
Collapse
Affiliation(s)
- Hong Yu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Hiroshi Nishio
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Joseph Barbi
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Marisa Mitchell-Flack
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Paolo D A Vignali
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Ying Zheng
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Andriana Lebid
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Kwang-Yu Chang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Juan Fu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Makenzie Higgins
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Ching-Tai Huang
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Xuehong Zhang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Zhiguang Li
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Lee Blosser
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Ada Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Charles Drake
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Drew Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
5
|
Silvestri B, Mochi M, Mawrie D, de Turris V, Colantoni A, Borhy B, Medici M, Anderson EN, Garone MG, Zammerilla CP, Simula M, Ballarino M, Pandey UB, Rosa A. HuD impairs neuromuscular junctions and induces apoptosis in human iPSC and Drosophila ALS models. Nat Commun 2024; 15:9618. [PMID: 39511225 PMCID: PMC11544248 DOI: 10.1038/s41467-024-54004-8] [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: 08/08/2023] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Defects at the neuromuscular junction (NMJ) are among the earliest hallmarks of amyotrophic lateral sclerosis (ALS). According to the "dying-back" hypothesis, NMJ disruption not only precedes but also triggers the subsequent degeneration of motoneurons in both sporadic (sALS) and familial (fALS) ALS. Using human induced pluripotent stem cells (iPSCs), we show that the RNA-binding protein HuD (ELAVL4) contributes to NMJ defects and apoptosis in FUS-ALS. HuD overexpression mimics the severe FUSP525L mutation, while its knockdown rescues the FUSP525L phenotypes. In Drosophila, neuronal overexpression of the HuD ortholog, elav, induces motor dysfunction, and its knockdown improves motor function in a FUS-ALS model. Finally, we report increased HuD levels upon oxidative stress in human motoneurons and in sALS patients with an oxidative stress signature. Based on these findings, we propose that HuD plays a role downstream of FUS mutations in fALS and in sALS related to oxidative stress.
Collapse
Affiliation(s)
- Beatrice Silvestri
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Michela Mochi
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Darilang Mawrie
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Valeria de Turris
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Alessio Colantoni
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Beatrice Borhy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Margherita Medici
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Eric Nathaniel Anderson
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Maria Giovanna Garone
- Stem Cell Medicine Department, Murdoch Children's Research Institute, Parkville, VIC, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW Melbourne, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Christopher Patrick Zammerilla
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Marco Simula
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Monica Ballarino
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Udai Bhan Pandey
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Alessandro Rosa
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy.
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy.
| |
Collapse
|
6
|
Meng P, Zhu L, Guo J, Li Y, Wei Y, Sun J, Zhu J. Preparation of recombinant neuritin protein. Protein Expr Purif 2024; 223:106554. [PMID: 39002828 DOI: 10.1016/j.pep.2024.106554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Neuritin plays an important role in promoting nerve injury repair and maintaining synaptic plasticity, making it a potential therapeutic target for the treatment of nerve injury and neurodegenerative diseases. The present study aimed to obtain an active, unlabeled neuritin protein. Initially, a neuritin protein expression system with an enterokinase site was constructed in Escherichia coli. After optimizing induction conditions and screening for high expression, a neuritin recombinant protein with purity exceeding 85 % was obtained through Ni-affinity chromatography. Subsequently, unlabeled neuritin with a molecular weight of 11 kDa was obtained through the enzymatic cleavage of the His label using an enterokinase. Furthermore, a neuritin recombinant protein with purity exceeding 95 % was obtained using gel chromatography. Functional investigations revealed that neurite outgrowth of PC12 cells was stimulated by the isolated neuritin. This study establishes a method to obtain active and unlabeled neuritin protein, providing a foundation for subsequent research on its biological functions.
Collapse
Affiliation(s)
- Pingping Meng
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Liyan Zhu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Jiatong Guo
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Yuanyuan Li
- Shawan City People's Hospital, Shawan, Xinjiang, 832100, China
| | - Yu Wei
- The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Jiawei Sun
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China.
| |
Collapse
|
7
|
Shimada T, Kohyama K, Yoshida T, Yamagata K. Neuritin Controls Axonal Branching in Serotonin Neurons: A Possible Mediator Involved in the Regulation of Depressive and Anxiety Behaviors via FGF Signaling. J Neurosci 2024; 44:e0129232024. [PMID: 39197941 PMCID: PMC11466069 DOI: 10.1523/jneurosci.0129-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
Abnormal neuronal morphological features, such as dendrite branching, axonal branching, and spine density, are thought to contribute to the symptoms of depression and anxiety. However, the role and molecular mechanisms of aberrant neuronal morphology in the regulation of mood disorders remain poorly characterized. Here, we show that neuritin, an activity-dependent protein, regulates the axonal morphology of serotonin neurons. Male neuritin knock-out (KO) mice harbored impaired axonal branches of serotonin neurons in the medial prefrontal cortex and basolateral region of the amygdala (BLA), and male neuritin KO mice exhibited depressive and anxiety-like behaviors. We also observed that the expression of neuritin was decreased by unpredictable chronic stress in the male mouse brain and that decreased expression of neuritin was associated with reduced axonal branching of serotonin neurons in the brain and with depressive and anxiety behaviors in mice. Furthermore, the stress-mediated impairments in axonal branching and depressive behaviors were reversed by the overexpression of neuritin in the BLA. The ability of neuritin to increase axonal branching in serotonin neurons involves fibroblast growth factor (FGF) signaling, and neuritin contributes to FGF-2-mediated axonal branching regulation in vitro. Finally, the oral administration of an FGF inhibitor reduced the axonal branching of serotonin neurons in the brain and caused depressive and anxiety behaviors in male mice. Our results support the involvement of neuritin in models of stress-induced depression and suggest that neuronal morphological plasticity may play a role in controlling animal behavior.
Collapse
Affiliation(s)
- Tadayuki Shimada
- Child Brain Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| | - Kuniko Kohyama
- Child Brain Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| | - Tomoyuki Yoshida
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Kanato Yamagata
- Child Brain Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
- Department of Psychiatry, Takada Nishishiro Hospital, Joetsu, Niigata 943-0834, Japan
| |
Collapse
|
8
|
Yu H, Nishio H, Barbi J, Mitchell-Flack M, Vignali PDA, Zheng Y, Lebid A, Chang KY, Fu J, Higgins M, Huang CT, Zhang X, Li Z, Blosser L, Tam A, Drake CG, Pardoll DM. Neurotrophic factor Neuritin modulates T cell electrical and metabolic state for the balance of tolerance and immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578284. [PMID: 38352414 PMCID: PMC10862906 DOI: 10.1101/2024.01.31.578284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The adaptive T cell response is accompanied by continuous rewiring of the T cell's electric and metabolic state. Ion channels and nutrient transporters integrate bioelectric and biochemical signals from the environment, setting cellular electric and metabolic states. Divergent electric and metabolic states contribute to T cell immunity or tolerance. Here, we report that neuritin (Nrn1) contributes to tolerance development by modulating regulatory and effector T cell function. Nrn1 expression in regulatory T cells promotes its expansion and suppression function, while expression in the T effector cell dampens its inflammatory response. Nrn1 deficiency causes dysregulation of ion channel and nutrient transporter expression in Treg and effector T cells, resulting in divergent metabolic outcomes and impacting autoimmune disease progression and recovery. These findings identify a novel immune function of the neurotrophic factor Nrn1 in regulating the T cell metabolic state in a cell context-dependent manner and modulating the outcome of an immune response.
Collapse
Affiliation(s)
- Hong Yu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hiroshi Nishio
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Joseph Barbi
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY14263, USA
| | - Marisa Mitchell-Flack
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Paolo D A Vignali
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: University of Pittsburgh, Carnegie Mellon
| | - Ying Zheng
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andriana Lebid
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kwang-Yu Chang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Juan Fu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Makenzie Higgins
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ching-Tai Huang
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: Infectious Diseases, Department of Medicine, Chang Gung Memorial Hospital, Taiwan
| | - Xuehong Zhang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian 116044, China
| | - Zhiguang Li
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian 116044, China
| | - Lee Blosser
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ada Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles G Drake
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| | - Drew M Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
Vasudevan A, Koushika SP. Physical presence of chemical synapses is necessary for turning behavior of anterograde synaptic vesicles at the branch point of PLM neurons in C. elegans. MICROPUBLICATION BIOLOGY 2024; 2024. [PMID: 38989012 PMCID: PMC11234195 DOI: 10.17912/micropub.biology.001204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024]
Abstract
Neurons exhibit complex branched axonal morphologies in both vertebrate and invertebrate systems, and show heterogeneity in the distribution of synaptic cargo across multiple synapses. It is possible that differences in transport across multiple branches contribute to the heterogeneity in cargo distribution across multiple synapses. However, the regulation of transport at axonal branch points is not well understood. We demonstrate that branch-specific transport of synaptic vesicles is dependent on the existence of a connection between the branch and synapses. The loss of this connection causes an immediate decrease in branch-specific transport of synaptic vesicles in the PLM neuron of C. elegans .
Collapse
Affiliation(s)
- Amruta Vasudevan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| |
Collapse
|
10
|
Lanfranchi M, Yandiev S, Meyer-Dilhet G, Ellouze S, Kerkhofs M, Dos Reis R, Garcia A, Blondet C, Amar A, Kneppers A, Polvèche H, Plassard D, Foretz M, Viollet B, Sakamoto K, Mounier R, Bourgeois CF, Raineteau O, Goillot E, Courchet J. The AMPK-related kinase NUAK1 controls cortical axons branching by locally modulating mitochondrial metabolic functions. Nat Commun 2024; 15:2487. [PMID: 38514619 PMCID: PMC10958033 DOI: 10.1038/s41467-024-46146-6] [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: 03/25/2023] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
The cellular mechanisms underlying axonal morphogenesis are essential to the formation of functional neuronal networks. We previously identified the autism-linked kinase NUAK1 as a central regulator of axon branching through the control of mitochondria trafficking. However, (1) the relationship between mitochondrial position, function and axon branching and (2) the downstream effectors whereby NUAK1 regulates axon branching remain unknown. Here, we report that mitochondria recruitment to synaptic boutons supports collateral branches stabilization rather than formation in mouse cortical neurons. NUAK1 deficiency significantly impairs mitochondrial metabolism and axonal ATP concentration, and upregulation of mitochondrial function is sufficient to rescue axonal branching in NUAK1 null neurons in vitro and in vivo. Finally, we found that NUAK1 regulates axon branching through the mitochondria-targeted microprotein BRAWNIN. Our results demonstrate that NUAK1 exerts a dual function during axon branching through its ability to control mitochondrial distribution and metabolic activity.
Collapse
Affiliation(s)
- Marine Lanfranchi
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Sozerko Yandiev
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Géraldine Meyer-Dilhet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Salma Ellouze
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500, Bron, France
| | - Martijn Kerkhofs
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Raphael Dos Reis
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Audrey Garcia
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Camille Blondet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Alizée Amar
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Anita Kneppers
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Hélène Polvèche
- Laboratoire de Biologie et Modelisation de la Cellule, Ecole Normale Superieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Universite Claude Bernard Lyon 1, 46 allée d'Italie F-69364, Lyon, France
- CECS/AFM, I-STEM, 28 rue Henri Desbruères, F-91100, Corbeil-Essonnes, France
| | - Damien Plassard
- CNRS UMR 7104, INSERM U1258, GenomEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Marc Foretz
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Benoit Viollet
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Rémi Mounier
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Cyril F Bourgeois
- Laboratoire de Biologie et Modelisation de la Cellule, Ecole Normale Superieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Universite Claude Bernard Lyon 1, 46 allée d'Italie F-69364, Lyon, France
| | - Olivier Raineteau
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500, Bron, France
| | - Evelyne Goillot
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Julien Courchet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France.
| |
Collapse
|
11
|
Silvestri B, Mochi M, Mawrie D, de Turris V, Colantoni A, Borhy B, Medici M, Anderson EN, Garone MG, Zammerilla CP, Pandey UB, Rosa A. HuD (ELAVL4) gain-of-function impairs neuromuscular junctions and induces apoptosis in in vitro and in vivo models of amyotrophic lateral sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.22.554258. [PMID: 38464028 PMCID: PMC10925158 DOI: 10.1101/2023.08.22.554258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Early defects at the neuromuscular junction (NMJ) are among the first hallmarks of the progressive neurodegenerative disease amyotrophic lateral sclerosis (ALS). According to the "dying back" hypothesis, disruption of the NMJ not only precedes, but is also a trigger for the subsequent degeneration of the motoneuron in both sporadic and familial ALS, including ALS caused by the severe FUS pathogenic variant P525L. However, the mechanisms linking genetic and environmental factors to NMJ defects remain elusive. By taking advantage of co-cultures of motoneurons and skeletal muscle derived from human induced pluripotent stem cells (iPSCs), we show that the neural RNA binding protein HuD (ELAVL4) may underlie NMJ defects and apoptosis in FUS-ALS. HuD overexpression in motoneurons phenocopies the severe FUSP525L mutation, while HuD knockdown in FUSP525L co-cultures produces phenotypic rescue. We validated these findings in vivo in a Drosophila FUS-ALS model. Neuronal-restricted overexpression of the HuD-related gene, elav, produces per se a motor phenotype, while neuronal-restricted elav knockdown significantly rescues motor dysfunction caused by FUS. Finally, we show that HuD levels increase upon oxidative stress in human motoneurons and in sporadic ALS patients with an oxidative stress signature. On these bases, we propose HuD as an important player downstream of FUS mutation in familial ALS, with potential implications for sporadic ALS related to oxidative stress.
Collapse
Affiliation(s)
- Beatrice Silvestri
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Center for Life Nano-& Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Michela Mochi
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Darilang Mawrie
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, PA, USA
| | - Valeria de Turris
- Center for Life Nano-& Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Alessio Colantoni
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Center for Life Nano-& Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Beatrice Borhy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Margherita Medici
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Eric Nathaniel Anderson
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, PA, USA
| | - Maria Giovanna Garone
- Department of Stem Cell Biology, Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Melbourne, Victoria, Australia
| | | | - Udai Bhan Pandey
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, PA, USA
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA USA
| | - Alessandro Rosa
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Center for Life Nano-& Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| |
Collapse
|
12
|
Meng P, Wei Y, Liang M, Yuan W, Zhu L, Sun J, Huang J, Zhu J. Fusion with CTP increases the stability of recombinant neuritin. Protein Expr Purif 2023; 212:106344. [PMID: 37567400 DOI: 10.1016/j.pep.2023.106344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/25/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Neuritin is a vital neurotrophin that plays an essential role in recovery from nerve injury and neurodegenerative diseases and may become a new target for treating these conditions. However, improving neuritin protein stability is an urgent problem. In this study, to obtain active and stable neuritin proteins, we added a carboxyl-terminal peptide (CTP) sequence containing four O-linked glycosylation sites to the C-terminus of neuritin and cloned it into the Chinese hamster ovary (CHO) expression system. The neuritin-CTP protein was purified using a His-Tag purification strategy after G418 screening of stable high-expression cell lines. Ultimately, we obtained neuritin-CTP protein with a purity >90%. Functional analyses showed that the purified neuritin-CTP protein promoted the neurite outgrowth of PC12 cells, and stability experiments showed that neuritin stability was increased by adding CTP. These results indicate that neuritin protein-CTP fusion effectively increases stability without affecting secretion and activity. This study offers a sound strategy for improving the stability of neuritin protein and provides material conditions for further study of the function of neuritin.
Collapse
Affiliation(s)
- Pingping Meng
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Yu Wei
- The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Mengjie Liang
- Department of Clinical Laboratory, Hospital of Xinjiang Production and Construction Corps/Second Affiliated Hospital, Medical School of Shihezi University, Urumqi, Xinjiang, 832000, China
| | - Wumei Yuan
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Liyan Zhu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Jiawei Sun
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China.
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China.
| |
Collapse
|
13
|
Zhu J, Li Y, Zhong C, Zhu M, Zheng Y, Xiong A, Meng P, Shan L, Li Y, Huang J. Neuritin affects the activity of neuralized-like 1 by promoting degradation and weakening its affinity for substrate. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1650-1658. [PMID: 37249336 PMCID: PMC10577452 DOI: 10.3724/abbs.2023098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
Neuritin plays a key role in neural development and regeneration by promoting neurite outgrowth and synapse maturation. Our previous research revealed the mechanism by which neuritin inhibits Notch signaling through interaction with neuralized-like 1 (Neurl1) to promote neurite growth. However, how neuritin regulates Notch signaling through Neurl1 has not been elucidated. Here, we first confirm that neuritin is an upstream regulator of Neurl1 and inhibits Notch signaling through Neurl1. Neurl1 is an E3 ubiquitin ligase that can promote ubiquitination and endocytosis of the Notch1 ligand Jagged1. Therefore, we observe the effect of neuritin on the ligase activity of Neurl1. The results indicate that neuritin inhibits Neurl1 activity by reducing the ubiquitination level and endocytosis of the target protein Jagged1. Moreover, we find that decreased activity of Neurl1 results in reduced expression of Notch receptor Notch intracellular domain (NICD) and downstream target gene hairy and enhancer of split-1 ( HES1). Furthermore, we investigate how neuritin affects Neurl1 enzyme activity. The results show that neuritin not only weakens the affinity between Neurl1 and Jagged1 but also promotes the degradation of Neurl1 by the 26S proteasome pathway. Taken together, our results suggest that neuritin negatively regulates Notch signaling by inhibiting the activity of Neurl1, promoting the degradation of Neurl1 and weakening the affinity of Neurl1 for Jagged1. Our study clarifies the molecular mechanisms of neuritin in regulating the Notch signaling pathway and provides new clues about how neuritin mediates neural regeneration and plasticity.
Collapse
Affiliation(s)
- Jingling Zhu
- Department of Biochemistry and Molecular BiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Yu Li
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Chen Zhong
- the First Affiliated Hospital of Shihezi University School of MedicineShihezi832000China
| | - Meiyi Zhu
- the First Affiliated Hospital of Shihezi University School of MedicineShihezi832000China
| | - Yan Zheng
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Anying Xiong
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Pingping Meng
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Liya Shan
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Yang Li
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Jin Huang
- Department of Biochemistry and Molecular BiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| |
Collapse
|
14
|
Hurst C, Pugh DA, Abreha MH, Duong DM, Dammer EB, Bennett DA, Herskowitz JH, Seyfried NT. Integrated Proteomics to Understand the Role of Neuritin (NRN1) as a Mediator of Cognitive Resilience to Alzheimer's Disease. Mol Cell Proteomics 2023; 22:100542. [PMID: 37024090 PMCID: PMC10233303 DOI: 10.1016/j.mcpro.2023.100542] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/16/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023] Open
Abstract
The molecular mechanisms and pathways enabling certain individuals to remain cognitively normal despite high levels of Alzheimer's disease (AD) pathology remain incompletely understood. These cognitively normal people with AD pathology are described as preclinical or asymptomatic AD (AsymAD) and appear to exhibit cognitive resilience to the clinical manifestations of AD dementia. Here we present a comprehensive network-based approach from cases clinically and pathologically defined as asymptomatic AD to map resilience-associated pathways and extend mechanistic validation. Multiplex tandem mass tag MS (TMT-MS) proteomic data (n = 7787 proteins) was generated on brain tissue from Brodmann area 6 and Brodmann area 37 (n = 109 cases, n = 218 total samples) and evaluated by consensus weighted gene correlation network analysis. Notably, neuritin (NRN1), a neurotrophic factor previously linked to cognitive resilience, was identified as a hub protein in a module associated with synaptic biology. To validate the function of NRN1 with regard to the neurobiology of AD, we conducted microscopy and physiology experiments in a cellular model of AD. NRN1 provided dendritic spine resilience against amyloid-β (Aβ) and blocked Aβ-induced neuronal hyperexcitability in cultured neurons. To better understand the molecular mechanisms of resilience to Aβ provided by NRN1, we assessed how exogenous NRN1 alters the proteome by TMT-MS (n = 8238 proteins) of cultured neurons and integrated the results with the AD brain network. This revealed overlapping synapse-related biology that linked NRN1-induced changes in cultured neurons with human pathways associated with cognitive resilience. Collectively, this highlights the utility of integrating the proteome from the human brain and model systems to advance our understanding of resilience-promoting mechanisms and prioritize therapeutic targets that mediate resilience to AD.
Collapse
Affiliation(s)
- Cheyenne Hurst
- Department of Biochemistry, Emory School of Medicine, Emory Goizueta Alzheimer's Disease Research Center, Atlanta, Georgia, USA
| | - Derian A Pugh
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Measho H Abreha
- Department of Biochemistry, Emory School of Medicine, Emory Goizueta Alzheimer's Disease Research Center, Atlanta, Georgia, USA
| | - Duc M Duong
- Department of Biochemistry, Emory School of Medicine, Emory Goizueta Alzheimer's Disease Research Center, Atlanta, Georgia, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory School of Medicine, Emory Goizueta Alzheimer's Disease Research Center, Atlanta, Georgia, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Jeremy H Herskowitz
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA.
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory School of Medicine, Emory Goizueta Alzheimer's Disease Research Center, Atlanta, Georgia, USA.
| |
Collapse
|
15
|
Liu H, Caballero-Florán RN, Hergenreder T, Yang T, Hull JM, Pan G, Li R, Veling MW, Isom LL, Kwan KY, Huang ZJ, Fuerst PG, Jenkins PM, Ye B. DSCAM gene triplication causes excessive GABAergic synapses in the neocortex in Down syndrome mouse models. PLoS Biol 2023; 21:e3002078. [PMID: 37079499 PMCID: PMC10118173 DOI: 10.1371/journal.pbio.3002078] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/14/2023] [Indexed: 04/21/2023] Open
Abstract
Down syndrome (DS) is caused by the trisomy of human chromosome 21 (HSA21). A major challenge in DS research is to identify the HSA21 genes that cause specific symptoms. Down syndrome cell adhesion molecule (DSCAM) is encoded by a HSA21 gene. Previous studies have shown that the protein level of the Drosophila homolog of DSCAM determines the size of presynaptic terminals. However, whether the triplication of DSCAM contributes to presynaptic development in DS remains unknown. Here, we show that DSCAM levels regulate GABAergic synapses formed on neocortical pyramidal neurons (PyNs). In the Ts65Dn mouse model for DS, where DSCAM is overexpressed due to DSCAM triplication, GABAergic innervation of PyNs by basket and chandelier interneurons is increased. Genetic normalization of DSCAM expression rescues the excessive GABAergic innervations and the increased inhibition of PyNs. Conversely, loss of DSCAM impairs GABAergic synapse development and function. These findings demonstrate excessive GABAergic innervation and synaptic transmission in the neocortex of DS mouse models and identify DSCAM overexpression as the cause. They also implicate dysregulated DSCAM levels as a potential pathogenic driver in related neurological disorders.
Collapse
Affiliation(s)
- Hao Liu
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - René N. Caballero-Florán
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Ty Hergenreder
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Tao Yang
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jacob M. Hull
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Geng Pan
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ruonan Li
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Macy W. Veling
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Kenneth Y. Kwan
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Z. Josh Huang
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, North Carolina, United States of America
| | - Peter G. Fuerst
- University of Idaho, Department of Biological Sciences, Moscow, Idaho, United States of America
| | - Paul M. Jenkins
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Bing Ye
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
16
|
Silvestri B, Mochi M, Garone MG, Rosa A. Emerging Roles for the RNA-Binding Protein HuD (ELAVL4) in Nervous System Diseases. Int J Mol Sci 2022; 23:14606. [PMID: 36498933 PMCID: PMC9736382 DOI: 10.3390/ijms232314606] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
The main goal of this review is to provide an updated overview of the involvement of the RNA-binding protein (RBP) HuD, encoded by the ELAVL4 gene, in nervous system development, maintenance, and function, and its emerging role in nervous system diseases. A particular focus is on recent studies reporting altered HuD levels, or activity, in disease models and patients. Substantial evidence suggests HuD involvement in Parkinson's disease (PD), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS). Interestingly, while possible disease-causing mutations in the ELAVL4 gene remain elusive, a common theme in these diseases seems to be the altered regulation of HuD at multiple steps, including post-transcriptional and post-translational levels. In turn, the changed activity of HuD can have profound implications for its target transcripts, which are overly stabilized in case of HuD gain of function (as proposed in PD and ALS) or reduced in case of decreased HuD binding (as suggested by some studies in AD). Moreover, the recent discovery that HuD is a component of pathological cytoplasmic inclusion in both familial and sporadic ALS patients might help uncover the common molecular mechanisms underlying such complex diseases. We believe that deepening our understanding of the involvement of HuD in neurodegeneration could help developing new diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Beatrice Silvestri
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Michela Mochi
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Giovanna Garone
- Department of Stem Cell Biology, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Alessandro Rosa
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| |
Collapse
|
17
|
Cline HT. Imaging Structural and Functional Dynamics in Xenopus Neurons. Cold Spring Harb Protoc 2021; 2022:pdb.top106773. [PMID: 34531329 DOI: 10.1101/pdb.top106773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In vivo time-lapse imaging has been a fruitful approach to identify structural and functional changes in the Xenopus nervous system in tadpoles and adult frogs. Structural imaging studies have identified fundamental aspects of brain connectivity, development, plasticity, and disease and have been instrumental in elucidating mechanisms regulating these events in vivo. Similarly, assessment of nervous system function using dynamic changes in calcium signals as a proxy for neuronal activity has demonstrated principles of neuron and circuit function and principles of information organization and transfer within the brain of living animals. Because of its many advantages as an experimental system, use of Xenopus has often been at the forefront of developing these imaging methods for in vivo applications. Protocols for in vivo structural and functional imaging-including cellular labeling strategies, image collection, and image analysis-will expand the use of Xenopus to understand brain development, function, and plasticity.
Collapse
Affiliation(s)
- Hollis T Cline
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Center, La Jolla, California 92039, USA
| |
Collapse
|
18
|
Garone MG, Birsa N, Rosito M, Salaris F, Mochi M, de Turris V, Nair RR, Cunningham TJ, Fisher EMC, Morlando M, Fratta P, Rosa A. ALS-related FUS mutations alter axon growth in motoneurons and affect HuD/ELAVL4 and FMRP activity. Commun Biol 2021; 4:1025. [PMID: 34471224 PMCID: PMC8410767 DOI: 10.1038/s42003-021-02538-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 08/10/2021] [Indexed: 12/13/2022] Open
Abstract
Mutations in the RNA-binding protein (RBP) FUS have been genetically associated with the motoneuron disease amyotrophic lateral sclerosis (ALS). Using both human induced pluripotent stem cells and mouse models, we found that FUS-ALS causative mutations affect the activity of two relevant RBPs with important roles in neuronal RNA metabolism: HuD/ELAVL4 and FMRP. Mechanistically, mutant FUS leads to upregulation of HuD protein levels through competition with FMRP for HuD mRNA 3'UTR binding. In turn, increased HuD levels overly stabilize the transcript levels of its targets, NRN1 and GAP43. As a consequence, mutant FUS motoneurons show increased axon branching and growth upon injury, which could be rescued by dampening NRN1 levels. Since similar phenotypes have been previously described in SOD1 and TDP-43 mutant models, increased axonal growth and branching might represent broad early events in the pathogenesis of ALS.
Collapse
Affiliation(s)
- Maria Giovanna Garone
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Nicol Birsa
- UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Maria Rosito
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Federico Salaris
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Michela Mochi
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Valeria de Turris
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | | | | | | | - Mariangela Morlando
- Department of Pharmaceutical Sciences, "Department of Excellence 2018-2022", University of Perugia, Perugia, Italy
| | - Pietro Fratta
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Alessandro Rosa
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy.
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy.
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
19
|
Kandasamy LC, Tsukamoto M, Banov V, Tsetsegee S, Nagasawa Y, Kato M, Matsumoto N, Takeda J, Itohara S, Ogawa S, Young LJ, Zhang Q. Limb-clasping, cognitive deficit and increased vulnerability to kainic acid-induced seizures in neuronal glycosylphosphatidylinositol deficiency mouse models. Hum Mol Genet 2021; 30:758-770. [PMID: 33607654 PMCID: PMC8161520 DOI: 10.1093/hmg/ddab052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 11/26/2022] Open
Abstract
Posttranslational modification of a protein with glycosylphosphatidylinositol (GPI) is a conserved mechanism exists in all eukaryotes. Thus far, >150 human GPI-anchored proteins have been discovered and ~30 enzymes have been reported to be involved in the biosynthesis and maturation of mammalian GPI. Phosphatidylinositol glycan biosynthesis class A protein (PIGA) catalyzes the very first step of GPI anchor biosynthesis. Patients carrying a mutation of the PIGA gene usually suffer from inherited glycosylphosphatidylinositol deficiency (IGD) with intractable epilepsy and intellectual developmental disorder. We generated three mouse models with PIGA deficits specifically in telencephalon excitatory neurons (Ex-M-cko), inhibitory neurons (In-M-cko) or thalamic neurons (Th-H-cko), respectively. Both Ex-M-cko and In-M-cko mice showed impaired long-term fear memory and were more susceptible to kainic acid-induced seizures. In addition, In-M-cko demonstrated a severe limb-clasping phenotype. Hippocampal synapse changes were observed in Ex-M-cko mice. Our Piga conditional knockout mouse models provide powerful tools to understand the cell-type specific mechanisms underlying inherited GPI deficiency and to test different therapeutic modalities.
Collapse
Affiliation(s)
- Lenin C Kandasamy
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Mina Tsukamoto
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Vitaliy Banov
- Laboratory for Behavioral Genetics, CBS, RIKEN, Wako 351-0198, Japan.,Institute of Neuroinformatics, University of Zürich, ETH Zürich, Zürich 8057, Switzerland
| | - Sambuu Tsetsegee
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Yutaro Nagasawa
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo 142-8555, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Junji Takeda
- Yabumoto Department of Intractable Disease Research, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | | | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Larry J Young
- Faculty of Human Sciences, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan.,Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University, Atlanta GA 30329, USA
| | - Qi Zhang
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan.,Laboratory for Behavioral Genetics, CBS, RIKEN, Wako 351-0198, Japan.,Faculty of Human Sciences, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
20
|
Oprişoreanu AM, Smith HL, Arya S, Webster R, Zhong Z, Eaton-Hart C, Wehner D, Cardozo MJ, Becker T, Talbot K, Becker CG. Interaction of Axonal Chondrolectin with Collagen XIXa1 Is Necessary for Precise Neuromuscular Junction Formation. Cell Rep 2020; 29:1082-1098.e10. [PMID: 31665626 DOI: 10.1016/j.celrep.2019.09.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/13/2019] [Accepted: 09/12/2019] [Indexed: 01/05/2023] Open
Abstract
Chondrolectin (Chodl) is needed for motor axon extension in zebrafish and is dysregulated in mouse models of spinal muscular atrophy (SMA). However, the mechanistic basis of Chodl function is not known. Here, we use Chodl-deficient zebrafish and mouse mutants to show that the absence of Chodl leads to anatomical and functional defects of the neuromuscular synapse. In zebrafish, the growth of an identified motor axon beyond an "en passant" synapse and later axon branching from synaptic points are impaired, leading to functional deficits. Mechanistically, motor-neuron-autonomous Chodl function depends on its intracellular domain and on binding muscle-derived collagen XIXa1 by its extracellular C-type lectin domain. Our data support evolutionarily conserved roles of Chodl in synaptogenesis and provide evidence for a "synapse-first" scenario of motor axon growth in zebrafish.
Collapse
Affiliation(s)
- Ana-Maria Oprişoreanu
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Hannah L Smith
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Sukrat Arya
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Richard Webster
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Zhen Zhong
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Charlotte Eaton-Hart
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Daniel Wehner
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcos J Cardozo
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK.
| | - Catherina G Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
21
|
Huang T, Li H, Zhang S, Liu F, Wang D, Xu J. Nrn1 Overexpression Attenuates Retinal Ganglion Cell Apoptosis, Promotes Axonal Regeneration, and Improves Visual Function Following Optic Nerve Crush in Rats. J Mol Neurosci 2020; 71:66-79. [DOI: 10.1007/s12031-020-01627-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022]
|
22
|
Song D, Li G, Hong Y, Zhang P, Zhu J, Yang L, Huang J. miR‑199a decreases Neuritin expression involved in the development of Alzheimer's disease in APP/PS1 mice. Int J Mol Med 2020; 46:384-396. [PMID: 32626916 PMCID: PMC7255456 DOI: 10.3892/ijmm.2020.4602] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/23/2020] [Indexed: 01/16/2023] Open
Abstract
Neuritin plays an important role in neural development and plasticity. A recent study demonstrated that increasing Neuritin levels attenuated synaptic damage in mice with Alzheimer's disease (AD), which exhibit a decreased Neuritin expression. However, it remains unclear as to whether Neuritin expression is regulated by microRNAs (miRNAs or miRs) in AD. In the present study, it was found that miR-199a decreased Neuritin expression and was therefore involved in the development of AD. Subsequently, differentially expressed miRNAs in AD from datasets and the literature were recruited, and those that could bind Neuritin were predicted using bioinformatics analysis. The present study then focused on the candidate miRNAs that were highly associated with Neuritin and were upregulated in AD. The expression patterns of the candidate miRNAs and Neuritin in the hippocampus and cortex of APP/PS1 (AD model) mice at different stages were then detected and analyzed. It was found that miR-199a expression was significantly increased in the early stages of AD and was negatively associated with Neuritin expression. Furthermore, it was revealed that the decreased Neuritin expression was due to the direct targeting of the Neuritin 3′-UTR by miR-199a. Finally, the association between the spatial memory capacity of APP/PS1 mice and the changes in miR-199a and Neuritin expression protein was investigated. On the whole, the data of the present study suggest that miR-199a is involved in the development of AD by regulating Neuritin expression.
Collapse
Affiliation(s)
- Dandan Song
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Guoxiang Li
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Yu Hong
- Department of Prevention Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310036, P.R. China
| | - Pan Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Lei Yang
- Department of Prevention Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310036, P.R. China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
23
|
Abstract
Antisense morpholino oligonucleotides (MOs) have become a valuable method to knockdown protein levels, to block with mRNA splicing and to interfere with miRNA function. MOs are widely used to alter gene expression in development of Xenopus and Zebrafish, where they are typically injected into the fertilized egg or blastomeres. Here we present methods to use electroporation to target delivery of MOs to the central nervous system of Xenopus laevis or Xenopus tropicalis tadpoles. Briefly, MO electroporation is accomplished by injecting MO solution into the brain ventricle and driving the MOs into cells of the brain with current passing between 2 platinum plate electrodes, positioned on either side of the target brain area. The method is relatively straightforward and uses standard equipment found in many neuroscience labs. A major advantage of electroporation is that it allows spatial and temporal control of MO delivery and therefore knockdown. Co-electroporation of MOs with cell type-specific fluorescent protein expression plasmids allows morphological analysis of cellular phenotypes. Furthermore, co-electroporation of MOs with rescuing plasmids allows assessment of specificity of the knockdown and phenotypic outcome. By combining MO-mediated manipulations with sophisticated assays of neuronal function, such as electrophysiological recording, behavioral assays, or in vivo time-lapse imaging of neuronal development, the functions of specific proteins and miRNAs within the developing nervous system can be elucidated. These methods can be adapted to apply antisense morpholinos to study protein and RNA function in a variety of complex tissues.
Collapse
Affiliation(s)
| | - Hollis T Cline
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
24
|
Rahmawati E, Yang WCV, Lei YP, Maurya PK, Chen HW, Tzeng CR. Decreased Level of Neurotrophic Factor Neuritin 1 in Women with Ovarian Endometriosis after Receiving Gonadotropin-Releasing Hormone Agonist Treatment. Int J Mol Sci 2019; 20:E4352. [PMID: 31491902 PMCID: PMC6770869 DOI: 10.3390/ijms20184352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 08/31/2019] [Accepted: 09/03/2019] [Indexed: 12/19/2022] Open
Abstract
This study aimed to investigate the effect of gonadotropin-releasing hormone agonist (GnRHa) treatment on the expression of neuritin 1 (NRN1) in women with ovarian endometriosis. We collected tissues and serum from women with endometriosis treated with (n = 45) or without (n = 37) GnRHa. NRN1 mRNA and protein levels were measured using qPCR and Western blot. Immunolocalization of NRN1 in endometriotic tissues was examined using immunohistochemistry. In addition, a follow-up study was carried out to monitor the serum level of NRN1 in patients before and after GnRHa treatment. Both mRNA (p = 0.046) and protein (p = 0.0155) levels of NRN1 were significantly lower in endometriotic tissues from patients receiving GnRHa treatment compared to the untreated group. Both epithelial and stromal cells of endometriotic tissues from untreated women with endometriosis exhibited stronger staining of NRN1 but not in those who were treated with GnRHa. The follow-up study showed that the serum level of the NRN1 concentration decreased significantly from 1149 ± 192.3 to 379.2 ± 80.16 pg/mL after GnRHa treatment (p = 0.0098). The expression of NRN1 was significantly lower in women with ovarian endometriosis treated with GnRHa. These results suggest that NRN1 may be a biomarker response to the effect of GnRHa treatment for patients with ovarian endometriosis.
Collapse
Affiliation(s)
- Endah Rahmawati
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Obstetrics and Gynecology, Faculty of Medicine Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia.
| | - Wei-Chung Vivian Yang
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yen-Ping Lei
- Department of Obstetrics and Gynecology, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India.
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| | - Chii-Ruey Tzeng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Obstetrics and Gynecology, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| |
Collapse
|
25
|
Tian R, Gachechiladze MA, Ludwig CH, Laurie MT, Hong JY, Nathaniel D, Prabhu AV, Fernandopulle MS, Patel R, Abshari M, Ward ME, Kampmann M. CRISPR Interference-Based Platform for Multimodal Genetic Screens in Human iPSC-Derived Neurons. Neuron 2019; 104:239-255.e12. [PMID: 31422865 DOI: 10.1016/j.neuron.2019.07.014] [Citation(s) in RCA: 321] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/25/2019] [Accepted: 07/12/2019] [Indexed: 12/28/2022]
Abstract
CRISPR/Cas9-based functional genomics have transformed our ability to elucidate mammalian cell biology. However, most previous CRISPR-based screens were conducted in cancer cell lines rather than healthy, differentiated cells. Here, we describe a CRISPR interference (CRISPRi)-based platform for genetic screens in human neurons derived from induced pluripotent stem cells (iPSCs). We demonstrate robust and durable knockdown of endogenous genes in such neurons and present results from three complementary genetic screens. First, a survival-based screen revealed neuron-specific essential genes and genes that improved neuronal survival upon knockdown. Second, a screen with a single-cell transcriptomic readout uncovered several examples of genes whose knockdown had strikingly cell-type-specific consequences. Third, a longitudinal imaging screen detected distinct consequences of gene knockdown on neuronal morphology. Our results highlight the power of unbiased genetic screens in iPSC-derived differentiated cell types and provide a platform for systematic interrogation of normal and disease states of neurons. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Ruilin Tian
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Biophysics Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | | | - Connor H Ludwig
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Matthew T Laurie
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jason Y Hong
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Diane Nathaniel
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Anika V Prabhu
- National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | | | - Rajan Patel
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Mehrnoosh Abshari
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD 20892, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA.
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
26
|
Bissen D, Foss F, Acker-Palmer A. AMPA receptors and their minions: auxiliary proteins in AMPA receptor trafficking. Cell Mol Life Sci 2019; 76:2133-2169. [PMID: 30937469 PMCID: PMC6502786 DOI: 10.1007/s00018-019-03068-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/12/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022]
Abstract
To correctly transfer information, neuronal networks need to continuously adjust their synaptic strength to extrinsic stimuli. This ability, termed synaptic plasticity, is at the heart of their function and is, thus, tightly regulated. In glutamatergic neurons, synaptic strength is controlled by the number and function of AMPA receptors at the postsynapse, which mediate most of the fast excitatory transmission in the central nervous system. Their trafficking to, at, and from the synapse, is, therefore, a key mechanism underlying synaptic plasticity. Intensive research over the last 20 years has revealed the increasing importance of interacting proteins, which accompany AMPA receptors throughout their lifetime and help to refine the temporal and spatial modulation of their trafficking and function. In this review, we discuss the current knowledge about the roles of key partners in regulating AMPA receptor trafficking and focus especially on the movement between the intracellular, extrasynaptic, and synaptic pools. We examine their involvement not only in basal synaptic function, but also in Hebbian and homeostatic plasticity. Included in our review are well-established AMPA receptor interactants such as GRIP1 and PICK1, the classical auxiliary subunits TARP and CNIH, and the newest additions to AMPA receptor native complexes.
Collapse
Affiliation(s)
- Diane Bissen
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany
| | - Franziska Foss
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
27
|
Du XF, Xu B, Zhang Y, Chen MJ, Du JL. A transgenic zebrafish model for in vivo long-term imaging of retinotectal synaptogenesis. Sci Rep 2018; 8:14077. [PMID: 30232367 PMCID: PMC6145912 DOI: 10.1038/s41598-018-32409-y] [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: 03/14/2017] [Accepted: 09/04/2018] [Indexed: 01/07/2023] Open
Abstract
The retinotectal synapse in larval zebrafish, combined with live time-lapse imaging, provides an advantageous model for study of the development and remodelling of central synapses in vivo. In previous studies, these synapses were labelled by transient expression of fluorescence-tagged synaptic proteins, which resulted in the dramatic variation of labelling patterns in each larva. Here, using GAL4-Upstream Activating Sequence (GAL4-UAS) methodology, we generated stable transgenic lines, which express EGFP-tagged synaptophysin (a presynaptic protein) in retinal ganglion cells (RGCs), to reliably label the pre-synaptic site of retinotectal synapses. This tool avoids the variable labelling of RGCs that occurs in transient transgenic larvae. We obtained several stable transgenic lines that differ consistently in the number of labelled RGCs. Using stable lines that consistently had a single labelled RGC, we could trace synaptogenic dynamics on an individual RGC axonal arbor across different developmental stages. In the stable lines that consistently had multiple labelled RGCs, we could simultaneously monitor both pre- and post-synaptic compartments by combining transient labelling of post-synaptic sites on individual tectal neurons. These tools allowed us to investigate molecular events underlying synaptogenesis and found that the microRNA-132 (miR-132) is required for developmental synaptogenesis. Thus, these transgenic zebrafish stable lines provide appropriate tools for studying central synaptogenesis and underlying molecular mechanisms in intact vertebrate brain.
Collapse
Affiliation(s)
- Xu-Fei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.
| | - Bing Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China
| | - Yu Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing, 100049, China
| | - Min-Jia Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, Shanghai, 200031, China
| | - Jiu-Lin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China. .,School of Future Technology, University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing, 100049, China. .,School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, Shanghai, 200031, China.
| |
Collapse
|
28
|
Khalil B, Morderer D, Price PL, Liu F, Rossoll W. mRNP assembly, axonal transport, and local translation in neurodegenerative diseases. Brain Res 2018; 1693:75-91. [PMID: 29462608 PMCID: PMC5997521 DOI: 10.1016/j.brainres.2018.02.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/31/2018] [Accepted: 02/13/2018] [Indexed: 12/12/2022]
Abstract
The development, maturation, and maintenance of the mammalian nervous system rely on complex spatiotemporal patterns of gene expression. In neurons, this is achieved by the expression of differentially localized isoforms and specific sets of mRNA-binding proteins (mRBPs) that regulate RNA processing, mRNA trafficking, and local protein synthesis at remote sites within dendrites and axons. There is growing evidence that axons contain a specialized transcriptome and are endowed with the machinery that allows them to rapidly alter their local proteome via local translation and protein degradation. This enables axons to quickly respond to changes in their environment during development, and to facilitate axon regeneration and maintenance in adult organisms. Aside from providing autonomy to neuronal processes, local translation allows axons to send retrograde injury signals to the cell soma. In this review, we discuss evidence that disturbances in mRNP transport, granule assembly, axonal localization, and local translation contribute to pathology in various neurodegenerative diseases, including spinal muscular atrophy (SMA), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Bilal Khalil
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Dmytro Morderer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Phillip L Price
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA; Department of Cell Biology, Emory University, Atlanta, GA 30322 USA
| | - Feilin Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA; Eye Center, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Wilfried Rossoll
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA.
| |
Collapse
|
29
|
Beattie CE, Kolb SJ. Spinal muscular atrophy: Selective motor neuron loss and global defect in the assembly of ribonucleoproteins. Brain Res 2018; 1693:92-97. [DOI: 10.1016/j.brainres.2018.02.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/10/2018] [Accepted: 02/15/2018] [Indexed: 12/13/2022]
|
30
|
Monavarfeshani A, Stanton G, Van Name J, Su K, Mills WA, Swilling K, Kerr A, Huebschman NA, Su J, Fox MA. LRRTM1 underlies synaptic convergence in visual thalamus. eLife 2018; 7:e33498. [PMID: 29424692 PMCID: PMC5826289 DOI: 10.7554/elife.33498] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/08/2018] [Indexed: 11/13/2022] Open
Abstract
It has long been thought that the mammalian visual system is organized into parallel pathways, with incoming visual signals being parsed in the retina based on feature (e.g. color, contrast and motion) and then transmitted to the brain in unmixed, feature-specific channels. To faithfully convey feature-specific information from retina to cortex, thalamic relay cells must receive inputs from only a small number of functionally similar retinal ganglion cells. However, recent studies challenged this by revealing substantial levels of retinal convergence onto relay cells. Here, we sought to identify mechanisms responsible for the assembly of such convergence. Using an unbiased transcriptomics approach and targeted mutant mice, we discovered a critical role for the synaptic adhesion molecule Leucine Rich Repeat Transmembrane Neuronal 1 (LRRTM1) in the emergence of retinothalamic convergence. Importantly, LRRTM1 mutant mice display impairment in visual behaviors, suggesting a functional role of retinothalamic convergence in vision.
Collapse
Affiliation(s)
- Aboozar Monavarfeshani
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
- Department of Biological SciencesVirginia TechBlacksburgUnited States
| | - Gail Stanton
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
- Virginia Tech Carilion School of MedicineRoanokeUnited States
| | - Jonathan Van Name
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
| | - Kaiwen Su
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
| | - William A Mills
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
- Translational Biology, Medicine, and Health Graduate ProgramVirginia TechBlacksburgUnited States
| | - Kenya Swilling
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
| | - Alicia Kerr
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
- Translational Biology, Medicine, and Health Graduate ProgramVirginia TechBlacksburgUnited States
| | | | - Jianmin Su
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
| | - Michael A Fox
- Developmental and Translational Neurobiology CenterVirginia Tech Carilion Research InstituteRoanokeUnited States
- Department of Biological SciencesVirginia TechBlacksburgUnited States
- Virginia Tech Carilion School of MedicineRoanokeUnited States
| |
Collapse
|
31
|
Liu Q, Zhang H, Xu J, Zhao D. Neuritin provides neuroprotection against experimental traumatic brain injury in rats. Int J Neurosci 2018; 128:811-820. [PMID: 29334295 DOI: 10.1080/00207454.2018.1424155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Neuritin is a neurotrophic factor that regulates neural growth and development. However, the role of neuritin in alleviating TBI has not been investigated. METHODS In this study, Sprague Dawley rats (n = 144) weighing 300 ± 50 g were categorized into control, sham, TBI and TBI + neuritin groups. The neurological scores and the ultrastructure of cortical neurons, apoptotic cells and caspase-3 were measured by using Garcia scoring system, transmission electron microscopy, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling, Western blot analysis and real-time RT-PCR at various time points post-TBI. CONCLUSIONS Our findings indicated that neuritin plays a protective role in TBI by improving neurological scores, repairing injured neurons and protecting the cortical neurons against apoptosis through inhibition of caspase-3 expression. Further investigation of the molecular mechanisms underlying caspase-3 inhibition by neuritin will provide a research avenue for potential TBI therapeutics.
Collapse
Affiliation(s)
- Qi Liu
- a Department of Neurosurgery , First Affiliated Hospital of Medical College, Shihezi University , Shihezi , Xinjiang , China.,b The Key Laboratory of Xinjiang Endemic and Ethnic Diseases , Medical College of Shihezi University , Shihezi , Xinjiang , China
| | - Hang Zhang
- a Department of Neurosurgery , First Affiliated Hospital of Medical College, Shihezi University , Shihezi , Xinjiang , China.,b The Key Laboratory of Xinjiang Endemic and Ethnic Diseases , Medical College of Shihezi University , Shihezi , Xinjiang , China
| | - Jian Xu
- a Department of Neurosurgery , First Affiliated Hospital of Medical College, Shihezi University , Shihezi , Xinjiang , China.,b The Key Laboratory of Xinjiang Endemic and Ethnic Diseases , Medical College of Shihezi University , Shihezi , Xinjiang , China
| | - Dong Zhao
- a Department of Neurosurgery , First Affiliated Hospital of Medical College, Shihezi University , Shihezi , Xinjiang , China.,b The Key Laboratory of Xinjiang Endemic and Ethnic Diseases , Medical College of Shihezi University , Shihezi , Xinjiang , China
| |
Collapse
|
32
|
Abstract
During development, the environment exerts a profound influence on the wiring of brain circuits. Due to the limited resolution of studies in fixed tissue, this experience-dependent structural plasticity was once thought to be restricted to a specific developmental time window. The recent introduction of two-photon microscopy for in vivo imaging has opened the door to repeated monitoring of individual neurons and the study of structural plasticity mechanisms at a very fine scale. In this review, we focus on recent work showing that synaptic structural rearrangements are a key mechanism mediating neural circuit adaptation and behavioral plasticity in the adult brain. We examine this work in the context of classic studies in the visual systems of model organisms, which have laid much of the groundwork for our understanding of activity-dependent synaptic remodeling and its role in brain plasticity.
Collapse
Affiliation(s)
- Kalen P Berry
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; .,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Elly Nedivi
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; .,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
33
|
Zhang S, Huang Y, Zhu J, Shan L, Gao J, Zhang Y, Yu N, Yang L, Huang J. Expression of hNeuritin protein in a baculovirus expression system and the analysis of its activity. Gene 2018; 647:129-135. [PMID: 29320757 DOI: 10.1016/j.gene.2018.01.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/04/2018] [Accepted: 01/06/2018] [Indexed: 12/11/2022]
Abstract
Neuritin plays an important role in the development and regeneration of the nervous system, and shows good prospects in the treatment and protection of the nervous system. To characterize neuritin function, we constructed a baculovirus expression system of neuritin, and identified the biological activity of the neuritin protein. The results and showed that the expression product could promote the neurite growth of dorsal root ganglion in chicken embryos. The neuritin open reading frame was amplified and cloned into the plasmid pFastBac™HTA. The pFastBac™HTA-neuritin was confirmed to be correct by PCR and DNA sequencing, and then transformed into Escherichia coli DH10Bac. The high purity recombinant Bacmid-neuritin (shuttle vectors) was obtained from DH10Bac through screening and identification. Recombinant virus, including the neuritin gene (virus-neuritin), was produced by transfection of SF9 cells using the bacmid-neuritin, and then amplified repeatedly to express the neuritin fusion protein. Finally, we identified the fusion protein with SDS-PAGE and western blotting, and optimized the best expression time of the neuritin fusion protein. We also analyzed the activity of the expressed protein by dorsal root ganglion from chicken embryos.
Collapse
Affiliation(s)
- Shuai Zhang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, China
| | - Yanhong Huang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, China
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, China
| | - Liya Shan
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, China
| | - Jianfeng Gao
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, China
| | - Yunhua Zhang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, China
| | - Na Yu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, China
| | - Lei Yang
- Hangzhou Normal University, Hangzhou, Zhejiang 310036, China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, China.
| |
Collapse
|
34
|
Borodinsky LN. Xenopus laevis as a Model Organism for the Study of Spinal Cord Formation, Development, Function and Regeneration. Front Neural Circuits 2017; 11:90. [PMID: 29218002 PMCID: PMC5704749 DOI: 10.3389/fncir.2017.00090] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/08/2017] [Indexed: 11/13/2022] Open
Abstract
The spinal cord is the first central nervous system structure to develop during vertebrate embryogenesis, underscoring its importance to the organism. Because of its early formation, accessibility to the developing spinal cord in amniotes is challenging, often invasive and the experimental approaches amenable to model systems like mammals are limited. In contrast, amphibians, in general and the African-clawed frog Xenopus laevis, in particular, offer model systems in which the formation of the spinal cord, the differentiation of spinal neurons and glia and the establishment of spinal neuron and neuromuscular synapses can be easily investigated with minimal perturbations to the whole organism. The significant advances on gene editing and microscopy along with the recent completion of the Xenopus laevis genome sequencing have reinvigorated the use of this classic model species to elucidate the mechanisms of spinal cord formation, development, function and regeneration.
Collapse
Affiliation(s)
- Laura N Borodinsky
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
35
|
In vivo imaging of Mauthner axon regeneration, remyelination and synapses re-establishment after laser axotomy in zebrafish larvae. Exp Neurol 2017; 300:67-73. [PMID: 29092800 DOI: 10.1016/j.expneurol.2017.10.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/24/2017] [Accepted: 10/27/2017] [Indexed: 12/31/2022]
Abstract
Zebrafish is an excellent model to study central nervous system (CNS) axonal degeneration and regeneration since we can observe these processes in vivo and in real time in transparent larvae. Previous studies have shown that Mauthner cell (M-cell) axon regenerates poorly after mechanical spinal cord injury. Inconsistent with this result, however, we have found that M-cell possesses a great capacity for axon regeneration after two-photon laser ablation. By using ZEISS LSM 710 two-photon microscope, we performed specific unilateral axotomy of GFP labeled M-cells in the Tol-056 enhancer trap line larvae. Our results showed that distal axons almost degenerated completely at 24h after laser axotomy. After that, the proximal axons initiated a robust regeneration and many of the M-cell axons almost regenerated fully at 4days post axotomy. Furthermore, we also visualized that regenerated axons were remyelinated when we severed fluorescent dye labeled M-cells in the Tg (mbp:EGFP-CAAX) line larvae. Moreover, by single M-cell co-electroporation with Syp:EGFP and DsRed2 plasmids we observed synapses re-establishment in vivo during laser injury-induced axon re-extension which suggested re-innervation of denervated pathways. In addition, we further demonstrated that nocodazole administration could completely abolish this regeneration capacity. These results together suggested that in vivo time-lapse imaging of M-cell axon laser injury may provide a powerful analytical model for understanding the underlying cellular and molecular mechanisms of the CNS axon regeneration.
Collapse
|
36
|
Zhang H, He X, Wang Y, Sun X, Zhu L, Lei C, Yin J, Li X, Hou F, He W, Zhao D. Neuritin attenuates early brain injury in rats after experimental subarachnoid hemorrhage. Int J Neurosci 2017; 127:1087-1095. [PMID: 28562156 DOI: 10.1080/00207454.2017.1337013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Hang Zhang
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Xuejun He
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Yezhong Wang
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Xiaokun Sun
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Licang Zhu
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Chao Lei
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Jiangwen Yin
- Department of Anesthesiology, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Xiaotian Li
- Department of Neurosurgery, Weifang People's HospitalWeifang, Shandong, China
| | - Fandi Hou
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Wengao He
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Dong Zhao
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| |
Collapse
|
37
|
Zhang P, Luo X, Guo Z, Xiong A, Dong H, Zhang Q, Liu C, Zhu J, Wang H, Yu N, Zhang J, Hong Y, Yang L, Huang J. Neuritin Inhibits Notch Signaling through Interacted with Neuralized to Promote the Neurite Growth. Front Mol Neurosci 2017. [PMID: 28642682 PMCID: PMC5462965 DOI: 10.3389/fnmol.2017.00179] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neuritin plays a key role in neural development and regeneration by promoting neurite outgrowth and synapse maturation. However, the mechanism of neuritin in modulating neurite growth has not been elucidated. Here, using yeast two-hybrid we screened and discovered the interaction of neuritin and neuralized (NEURL1), which is an important regulator that can activate Notch signaling through promoting endocytosis of Notch ligand. And then we identified the interaction of neuritin and neuralized by co-immunoprecipitation (IP) assays, and clarified that neuritin and NEURL1 were co-localized on the cell membrane of SH-SY5Y cells. Moreover, neuritin significantly suppressed Notch ligand Jagged1 (JAG1) endocytosis promoted by NEURL1, and then inhibited the activation of Notch receptor Notch intracellular domain (NICD) and decreased the expression of downstream gene hairy and enhancer of split-1 (HES1). Importantly, the effect of neuritin on inhibiting Notch signaling was rescued by NEURL1, which indicated that neuritin is an upstream and negative regulator of NEURL1 to inhibit Notch signaling through interaction with NEURL1. Notably, recombinant neuritin restored the retraction of neurites caused by activation of Notch, and neurite growth stimulated by neuritin was partially blocked by NEURL1. These findings establish neuritin as an upstream and negative regulator of NEURL1 that inhibits Notch signaling to promote neurite growth. This mechanism connects neuritin with Notch signaling, and provides a valuable foundation for further investigation of neuritin's role in neurodevelopment and neural plasticity.
Collapse
Affiliation(s)
- Pan Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Xing Luo
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Zheng Guo
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Anying Xiong
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Hongchang Dong
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Qiao Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Chunyan Liu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Haiyan Wang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Na Yu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Jinli Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Yu Hong
- School of Medicine, Hangzhou Normal UniversityHangzhou, China
| | - Lei Yang
- School of Medicine, Hangzhou Normal UniversityHangzhou, China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| |
Collapse
|
38
|
Lu JM, Liu DD, Li ZY, Ling C, Mei YA. Neuritin Enhances Synaptic Transmission in Medial Prefrontal Cortex in Mice by Increasing CaV3.3 Surface Expression. Cereb Cortex 2017; 27:3842-3855. [DOI: 10.1093/cercor/bhx082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 03/23/2017] [Indexed: 02/02/2023] Open
Affiliation(s)
- Jun-Mei Lu
- State Key Laboratory of Medical Neurobiology and School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Dong-Dong Liu
- State Key Laboratory of Medical Neurobiology and School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Zhao-Yang Li
- State Key Laboratory of Medical Neurobiology and School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Chen Ling
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan-Ai Mei
- State Key Laboratory of Medical Neurobiology and School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| |
Collapse
|
39
|
Edwards-Faret G, Muñoz R, Méndez-Olivos EE, Lee-Liu D, Tapia VS, Larraín J. Spinal cord regeneration in Xenopus laevis. Nat Protoc 2017; 12:372-389. [PMID: 28102835 DOI: 10.1038/nprot.2016.177] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Here we present a protocol for the husbandry of Xenopus laevis tadpoles and froglets, and procedures to study spinal cord regeneration. This includes methods to induce spinal cord injury (SCI); DNA and morpholino electroporation for genetic studies; in vivo imaging for cell analysis; a swimming test to measure functional recovery; and a convenient model for screening for new compounds that promote neural regeneration. These protocols establish X. laevis as a unique model organism for understanding spinal cord regeneration by comparing regenerative and nonregenerative stages. This protocol can be used to understand the molecular and cellular mechanisms involved in nervous system regeneration, including neural stem and progenitor cell (NSPC) proliferation and neurogenesis, extrinsic and intrinsic mechanisms involved in axon regeneration, glial response and scar formation, and trophic factors. For experienced personnel, husbandry takes 1-2 months; SCI can be achieved in 5-15 min; and swimming recovery takes 20-30 d.
Collapse
Affiliation(s)
- Gabriela Edwards-Faret
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rosana Muñoz
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Emilio E Méndez-Olivos
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Dasfne Lee-Liu
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Victor S Tapia
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Larraín
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
40
|
Soluble cpg15 from Astrocytes Ameliorates Neurite Outgrowth Recovery of Hippocampal Neurons after Mouse Cerebral Ischemia. J Neurosci 2017; 37:1628-1647. [PMID: 28069924 DOI: 10.1523/jneurosci.1611-16.2016] [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: 05/18/2016] [Revised: 12/22/2016] [Accepted: 12/30/2016] [Indexed: 11/21/2022] Open
Abstract
The present study focuses on the function of cpg15, a neurotrophic factor, in ischemic neuronal recovery using transient global cerebral ischemic (TGI) mouse model and oxygen-glucose deprivation (OGD)-treated primary cultured cells. The results showed that expression of cpg15 proteins in astrocytes, predominantly the soluble form, was significantly increased in mouse hippocampus after TGI and in the cultured astrocytes after OGD. Addition of the medium from the cpg15-overexpressed astrocytic culture into the OGD-treated hippocampal neuronal cultures reduces the neuronal injury, whereas the recovery of neurite outgrowths of OGD-injured neurons was prevented when cpg15 in the OGD-treated astrocytes was knocked down, or the OGD-treated-astrocytic medium was immunoadsorbed by cpg15 antibody. Furthermore, lentivirus-delivered knockdown of cpg15 expression in mouse hippocampal astrocytes diminishes the dendritic branches and exacerbates injury of neurons in CA1 region after TGI. In addition, treatment with inhibitors of MEK1/2, PI3K, and TrkA decreases, whereas overexpression of p-CREB, but not dp-CREB, increases the expression of cpg15 in U118 or primary cultured astrocytes. Also, it is observed that the Flag-tagged soluble cpg15 from the astrocytes transfected with Flag-tagged cpg15-expressing plasmids adheres to the surface of neuronal bodies and the neurites. In conclusion, our results suggest that the soluble cpg15 from astrocytes induced by ischemia could ameliorate the recovery of the ischemic-injured hippocampal neurons via adhering to the surface of neurons. The upregulated expression of cpg15 in astrocytes may be activated via MAPK and PI3K signal pathways, and regulation of CREB phosphorylation.SIGNIFICANCE STATEMENT Neuronal plasticity plays a crucial role in the amelioration of neurological recovery of ischemic injured brain, which remains a challenge for clinic treatment of cerebral ischemia. cpg15 as a synaptic plasticity-related factor may participate in promoting the recovery process; however, the underlying mechanisms are still largely unknown. The objective of this study is to reveal the function and mechanism of neuronal-specific cpg15 expressed in astrocytes after ischemia induction, in promoting the recovery of injured neurons. Our findings provided new mechanistic insight into the neurological recovery, which might help develop novel therapeutic options for cerebral ischemia via astrocytic-targeting interference of gene expression.
Collapse
|
41
|
Yener İH, Topaloglu H, Erdem-Özdamar S, Dayangac-Erden D. Transcript levels of plastin 3 and neuritin 1 modifier genes in spinal muscular atrophy siblings. Pediatr Int 2017; 59:53-56. [PMID: 27279027 DOI: 10.1111/ped.13052] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/20/2016] [Accepted: 06/02/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND In single gene disorders, patients with the same genotype may have variations in severity. One of the main factors affecting disease severity is modifier genes. Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder caused by degeneration of alpha motor neurons. Plastin 3 (PLS3) is a phenotypic modifier of SMA, and neuritin 1 (NRN1) has also been suggested as a possible modifier gene. The aim of the present study was therefore to analyze PLS3 and NRN1 expression in SMA siblings in four families. METHODS The study group consisted of four SMA families with seven with discordant phenotype and two affected siblings. Total RNA was isolated from whole blood. PLS3 and NRN1 expression was analyzed on quantitative real-time polymerase chain reaction. RESULTS In family 1 only NRN1 expression was increased in the mildly affected sister. In family 2 only PLS3 had a modifier effect. Family 3, which had type III siblings with identical clinical phenotypes, had similar PLS3 expression between the siblings but no NRN1 expression. In family 4, neither PLS3 nor NRN1 had any correlation with severity. CONCLUSION On analysis of the expression of NRN1 in SMA patients for the first time, NRN1 could be a potential modifier gene. PLS3 expression does not always modify SMA phenotype. In patients with no modifier effect of known genes, genome sequencing and transcriptome analysis are promising for the identification of novel modifiers and understanding of SMA pathophysiology.
Collapse
Affiliation(s)
- İnci Hande Yener
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Haluk Topaloglu
- Department of Pediatric Neurology, Faculty of Medicine, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Sevim Erdem-Özdamar
- Department of Neurology, Faculty of Medicine, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Didem Dayangac-Erden
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Sihhiye, Ankara, Turkey
| |
Collapse
|
42
|
Wang H, Li X, Shan L, Zhu J, Chen R, Li Y, Yuan W, Yang L, Huang J. Recombinant hNeuritin Promotes Structural and Functional Recovery of Sciatic Nerve Injury in Rats. Front Neurosci 2016; 10:589. [PMID: 28066172 PMCID: PMC5177646 DOI: 10.3389/fnins.2016.00589] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/08/2016] [Indexed: 01/29/2023] Open
Abstract
Neuritin is a new neurotropic factor implicated in nervous system development and plasticity. Studies have shown that Neuritin is upregulated in injured nerves, suggesting that it is involved in nerve repair. To test this hypothesis, we investigated whether recombinant human Neuritin could restore nerve structure and function in a rat model of sciatic nerve injury. Neuritin treatment had a dose-dependent effect on functional recovery 4 weeks after injury, as determined by the walking-track test. Similar trends were observed for gastrocnemius muscular strength and nerve conduction velocity. Additionally, sciatic nerve fiber density and organization as well as degree of remyelination were increased, while growth-associated protein 43 and neurofilament 200 expression was upregulated upon treatment with Neuritin. These findings demonstrate that Neuritin stimulates nerve regeneration and functional recovery and thus promotes the repair of injured sciatic nerves.
Collapse
Affiliation(s)
- Haiyan Wang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine Shihezi, China
| | - Xinli Li
- Laboratory Medicine Department of Sixth People's Hospital of Chengdu, Chengdu, China
| | - Liya Shan
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine Shihezi, China
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine Shihezi, China
| | - Rong Chen
- Occupational and Environmental Health, Department of Preventive Medicine, School of Medicine, Hangzhou Normal University Hangzhou, China
| | - Yuan Li
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Hangzhou Normal University Hangzhou, China
| | - Wumei Yuan
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine Shihezi, China
| | - Lei Yang
- Occupational and Environmental Health, Department of Preventive Medicine, School of Medicine, Hangzhou Normal University Hangzhou, China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine Shihezi, China
| |
Collapse
|
43
|
Leijon SC, Peyda S, Magnusson AK. Temporal processing capacity in auditory-deprived superior paraolivary neurons is rescued by sequential plasticity during early development. Neuroscience 2016; 337:315-330. [DOI: 10.1016/j.neuroscience.2016.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 09/01/2016] [Accepted: 09/09/2016] [Indexed: 01/04/2023]
|
44
|
Fatjó-Vilas M, Prats C, Pomarol-Clotet E, Lázaro L, Moreno C, González-Ortega I, Lera-Miguel S, Miret S, Muñoz MJ, Ibáñez I, Campanera S, Giralt-López M, Cuesta MJ, Peralta V, Ortet G, Parellada M, González-Pinto A, McKenna PJ, Fañanás L. Involvement of NRN1 gene in schizophrenia-spectrum and bipolar disorders and its impact on age at onset and cognitive functioning. World J Biol Psychiatry 2016; 17:129-39. [PMID: 26700405 DOI: 10.3109/15622975.2015.1093658] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Neuritin 1 gene (NRN1) is involved in neurodevelopment processes and synaptic plasticity and its expression is regulated by brain-derived neurotrophic factor (BDNF). We aimed to investigate the association of NRN1 with schizophrenia-spectrum disorders (SSD) and bipolar disorders (BPD), to explore its role in age at onset and cognitive functioning, and to test the epistasis between NRN1 and BDNF. METHODS The study was developed in a sample of 954 SSD/BPD patients and 668 healthy subjects. Genotyping analyses included 11 SNPs in NRN1 and one functional SNP in BDNF. RESULTS The frequency of the haplotype C-C (rs645649-rs582262) was significantly increased in patients compared to controls (P = 0.0043), while the haplotype T-C-C-T-C-A (rs3763180-rs10484320-rs4960155-rs9379002-rs9405890-rs1475157) was more frequent in controls (P = 3.1 × 10(-5)). The variability at NRN1 was nominally related to changes in age at onset and to differences in intelligence quotient, in SSD patients. Epistasis between NRN1 and BDNF was significantly associated with the risk for SSD/BPD (P = 0.005). CONCLUSIONS Results suggest that: (i) NRN1 variability is a shared risk factor for both SSD and BPD, (ii) NRN1 may have a selective impact on age at onset and intelligence in SSD, and (iii) the role of NRN1 seems to be not independent of BDNF.
Collapse
Affiliation(s)
- Mar Fatjó-Vilas
- a Departament de Biologia Animal, Facultat de Biologia, Universitat de Barcelona , Barcelona , Spain ; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain;,b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain
| | - Claudia Prats
- a Departament de Biologia Animal, Facultat de Biologia, Universitat de Barcelona , Barcelona , Spain ; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain;,b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain
| | - Edith Pomarol-Clotet
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,c FIDMAG Germanes Hospitalàries, Research Foundation , Barcelona , Spain
| | - Luisa Lázaro
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,d Servei de Psiquiatria i Psicologia Infantil i Juvenil, Hospital Clínic de Barcelona , Barcelona , Spain ;,e Institut d'investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain; Departament de Psiquiatria i Psicobiologia Clínica, Facultat de Medicina, Universitat de Barcelona , Barcelona , Spain
| | - Carmen Moreno
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,f Servicio de Psiquiatría del Niño y del Adolescente , Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Gregorio Marañón (IiSGM); Departamento de Psiquiatría, Facultad de Medicina, Universidad Complutense , Madrid , Spain
| | - Itxaso González-Ortega
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,g Psychiatry Service, University Hospital of Alava-Santiago, EMBREC, EHU/UPV University of the Basque Country, Kronikgune , Vitoria , Spain
| | - Sara Lera-Miguel
- d Servei de Psiquiatria i Psicologia Infantil i Juvenil, Hospital Clínic de Barcelona , Barcelona , Spain
| | - Salvador Miret
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,h Centre de Salut Mental d'Adults de Lleida, Servei de Psiquiatria, Salut Mental i Addiccions, Hospital Universitari Santa Maria de Lleida , Lleida , Spain
| | - Ma José Muñoz
- i Àrea d'Adolescents, Complex Assistencial en Salut Mental Benito Menni, Sant Boi De Llobregat , Spain
| | - Ignacio Ibáñez
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,j Departament de Psicologia Bàsica , Clínica i Psicobiologia, Facultat de Ciències de la Salut, Universitat Jaume I , Castelló , Spain
| | - Sílvia Campanera
- h Centre de Salut Mental d'Adults de Lleida, Servei de Psiquiatria, Salut Mental i Addiccions, Hospital Universitari Santa Maria de Lleida , Lleida , Spain
| | - Maria Giralt-López
- i Àrea d'Adolescents, Complex Assistencial en Salut Mental Benito Menni, Sant Boi De Llobregat , Spain
| | - Manuel J Cuesta
- k Servicio de Psiquiatría, Complejo Hospitalario de Navarra, Pamplona Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA) , Pamplona , Spain
| | - Victor Peralta
- k Servicio de Psiquiatría, Complejo Hospitalario de Navarra, Pamplona Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA) , Pamplona , Spain
| | - Generós Ortet
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,j Departament de Psicologia Bàsica , Clínica i Psicobiologia, Facultat de Ciències de la Salut, Universitat Jaume I , Castelló , Spain
| | - Mara Parellada
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,f Servicio de Psiquiatría del Niño y del Adolescente , Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Gregorio Marañón (IiSGM); Departamento de Psiquiatría, Facultad de Medicina, Universidad Complutense , Madrid , Spain
| | - Ana González-Pinto
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,g Psychiatry Service, University Hospital of Alava-Santiago, EMBREC, EHU/UPV University of the Basque Country, Kronikgune , Vitoria , Spain
| | - Peter J McKenna
- b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain ;,c FIDMAG Germanes Hospitalàries, Research Foundation , Barcelona , Spain
| | - Lourdes Fañanás
- a Departament de Biologia Animal, Facultat de Biologia, Universitat de Barcelona , Barcelona , Spain ; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain;,b Instituto De Salud Carlos III, Centro De Investigación Biomédica En Red De Salud Mental (CIBERSAM) , Madrid , Spain
| |
Collapse
|
45
|
Pratt KG, Hiramoto M, Cline HT. An Evolutionarily Conserved Mechanism for Activity-Dependent Visual Circuit Development. Front Neural Circuits 2016; 10:79. [PMID: 27818623 PMCID: PMC5073143 DOI: 10.3389/fncir.2016.00079] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/26/2016] [Indexed: 12/01/2022] Open
Abstract
Neural circuit development is an activity-dependent process. This activity can be spontaneous, such as the retinal waves that course across the mammalian embryonic retina, or it can be sensory-driven, such as the activation of retinal ganglion cells (RGCs) by visual stimuli. Whichever the source, neural activity provides essential instruction to the developing circuit. Indeed, experimentally altering activity has been shown to impact circuit development and function in many different ways and in many different model systems. In this review, we contemplate the idea that retinal waves in amniotes, the animals that develop either in ovo or utero (namely reptiles, birds and mammals) could be an evolutionary adaptation to life on land, and that the anamniotes, animals whose development is entirely external (namely the aquatic amphibians and fish), do not display retinal waves, most likely because they simply don’t need them. We then review what is known about the function of both retinal waves and visual stimuli on their respective downstream targets, and predict that the experience-dependent development of the tadpole visual system is a blueprint of what will be found in future studies of the effects of spontaneous retinal waves on instructing development of retinorecipient targets such as the superior colliculus (SC) and the lateral geniculate nucleus.
Collapse
Affiliation(s)
- Kara G Pratt
- Program in Neuroscience, Department of Zoology and Physiology, University of Wyoming Laramie, WY, USA
| | - Masaki Hiramoto
- Department of Molecular and Cellular Neuroscience and The Dorris Neuroscience Center, The Scripps Research Institute La Jolla, CA, USA
| | - Hollis T Cline
- Department of Molecular and Cellular Neuroscience and The Dorris Neuroscience Center, The Scripps Research Institute La Jolla, CA, USA
| |
Collapse
|
46
|
Neuritin Mediates Activity-Dependent Axonal Branch Formation in Part via FGF Signaling. J Neurosci 2016; 36:4534-48. [PMID: 27098696 DOI: 10.1523/jneurosci.1715-15.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 03/09/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Aberrant branch formation of granule cell axons (mossy fiber sprouting) is observed in the dentate gyrus of many patients with temporal lobe epilepsy and in animal models of epilepsy. However, the mechanisms underlying mossy fiber sprouting remain elusive. Based on the hypothesis that seizure-mediated gene expression induces abnormal mossy fiber growth, we screened activity-regulated genes in the hippocampus and found that neuritin, an extracellular protein anchored to the cell surface, was rapidly upregulated after electroconvulsive seizures. Overexpression of neuritin in the cultured rat granule cells promoted their axonal branching. Also, kainic acid-dependent axonal branching was abolished in the cultured granule cells fromneuritinknock-out mice, suggesting that neuritin may be involved in activity-dependent axonal branching. Moreover,neuritinknock-out mice showed less-severe seizures in chemical kindling probably by reduced mossy fiber sprouting and/or increased seizure resistance. We found that inhibition of the fibroblast growth factor (FGF) receptor attenuated the neuritin-dependent axonal branching. FGF administration also increased branching in granule neurons, whereasneuritinknock-out mice did not show FGF-dependent axonal branching. In addition, FGF and neuritin treatment enhanced the recruitment of FGF receptors to the cell surface. These findings suggest that neuritin and FGF cooperate in inducing mossy fiber sprouting through FGF signaling. Together, these results suggest that FGF and neuritin-mediated axonal branch induction are involved in the aggravation of epilepsy. SIGNIFICANCE STATEMENT This study reveals the molecular mechanism underlying mossy fiber sprouting. Mossy fiber sprouting is the aberrant axonal branching of granule neurons in the hippocampus, which is observed in patients with epilepsy. Excess amounts of neuritin, a protein upregulated by neural activity, promoted axonal branching in granule neurons. A deficiency of neuritin suppressed mossy fiber sprouting and resulted in mitigation of seizure severity. Neuritin and fibroblast growth factor (FGF) cooperated in stimulating FGF signaling and enhancing axonal branching. Neuritin is necessary for FGF-mediated recruitment of FGF receptors to the cell surface. The recruitment of FGF receptors would promote axonal branching. The discovery of this new mechanism should contribute to the development of novel antiepileptic drugs to inhibit axonal branching via neuritin-FGF signaling.
Collapse
|
47
|
Gao R, Li X, Xi S, Wang H, Zhang H, Zhu J, Shan L, Song X, Luo X, Yang L, Huang J. Exogenous Neuritin Promotes Nerve Regeneration After Acute Spinal Cord Injury in Rats. Hum Gene Ther 2016; 27:544-54. [DOI: 10.1089/hum.2015.159] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Rui Gao
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xingyi Li
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Shaosong Xi
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Haiyan Wang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Hong Zhang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Liya Shan
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiaoming Song
- School of Medicine & Health Management, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xing Luo
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Lei Yang
- School of Medicine & Health Management, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| |
Collapse
|
48
|
Yao JJ, Zhao QR, Liu DD, Chow CW, Mei YA. Neuritin Up-regulates Kv4.2 α-Subunit of Potassium Channel Expression and Affects Neuronal Excitability by Regulating the Calcium-Calcineurin-NFATc4 Signaling Pathway. J Biol Chem 2016; 291:17369-81. [PMID: 27307045 PMCID: PMC5016134 DOI: 10.1074/jbc.m115.708883] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Indexed: 11/06/2022] Open
Abstract
Neuritin is an important neurotrophin that regulates neural development, synaptic plasticity, and neuronal survival. Elucidating the downstream molecular signaling is important for potential therapeutic applications of neuritin in neuronal dysfunctions. We previously showed that neuritin up-regulates transient potassium outward current (IA) subunit Kv4.2 expression and increases IA densities, in part by activating the insulin receptor signaling pathway. Molecular mechanisms of neuritin-induced Kv4.2 expression remain elusive. Here, we report that the Ca(2+)/calcineurin (CaN)/nuclear factor of activated T-cells (NFAT) c4 axis is required for neuritin-induced Kv4.2 transcriptional expression and potentiation of IA densities in cerebellum granule neurons. We found that neuritin elevates intracellular Ca(2+) and increases Kv4.2 expression and IA densities; this effect was sensitive to CaN inhibition and was eliminated in Nfatc4(-/-) mice but not in Nfatc2(-/-) mice. Stimulation with neuritin significantly increased nuclear accumulation of NFATc4 in cerebellum granule cells and HeLa cells, which expressed IR. Furthermore, NFATc4 was recruited to the Kv4.2 gene promoter loci detected by luciferase reporter and chromatin immunoprecipitation assays. More importantly, data obtained from cortical neurons following adeno-associated virus-mediated overexpression of neuritin indicated that reduced neuronal excitability and increased formation of dendritic spines were abrogated in the Nfatc4(-/-) mice. Together, these data demonstrate an indispensable role for the CaN/NFATc4 signaling pathway in neuritin-regulated neuronal functions.
Collapse
Affiliation(s)
- Jin-Jing Yao
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Qian-Ru Zhao
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Dong-Dong Liu
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Chi-Wing Chow
- the Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York 10461
| | - Yan-Ai Mei
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| |
Collapse
|
49
|
Song MY, Tian FF, Dang J, Huang WJ, Guo JL. Possible Role of Protein CPG15 in Hippocampal Mossy Fiber Sprouting Under Conditions of Pentylenetetrazole Kindling. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9533-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
50
|
Zhao QR, Lu JM, Yao JJ, Zhang ZY, Ling C, Mei YA. Neuritin reverses deficits in murine novel object associative recognition memory caused by exposure to extremely low-frequency (50 Hz) electromagnetic fields. Sci Rep 2015; 5:11768. [PMID: 26138388 PMCID: PMC4650637 DOI: 10.1038/srep11768] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 06/01/2015] [Indexed: 12/21/2022] Open
Abstract
Animal studies have shown that electromagnetic field exposure may interfere with the activity of brain cells, thereby generating behavioral and cognitive disturbances. However, the underlying mechanisms and possible preventions are still unknown. In this study, we used a mouse model to examine the effects of exposure to extremely low-frequency (50 Hz) electromagnetic fields (ELF MFs) on a recognition memory task and morphological changes of hippocampal neurons. The data showed that ELF MFs exposure (1 mT, 12 h/day) induced a time-dependent deficit in novel object associative recognition memory and also decreased hippocampal dendritic spine density. This effect was observed without corresponding changes in spontaneous locomotor activity and was transient, which has only been seen after exposing mice to ELF MFs for 7-10 days. The over-expression of hippocampal neuritin, an activity-dependent neurotrophic factor, using an adeno-associated virus (AAV) vector significantly increased the neuritin level and dendritic spine density. This increase was paralleled with ELF MFs exposure-induced deficits in recognition memory and reductions of dendritic spine density. Collectively, our study provides evidence for the association between ELF MFs exposure, impairment of recognition memory, and resulting changes in hippocampal dendritic spine density. Neuritin prevented this ELF MFs-exposure-induced effect by increasing the hippocampal spine density.
Collapse
Affiliation(s)
- Qian-Ru Zhao
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Jun-Mei Lu
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Jin-Jing Yao
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Zheng-Yu Zhang
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Yan-Ai Mei
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| |
Collapse
|