1
|
Xie Y, Wang R, McClatchy DB, Ma Y, Diedrich J, Sanchez-Alavez M, Petrascheck M, Yates JR, Cline HT. Activity-dependent synthesis of Emerin gates neuronal plasticity by regulating proteostasis. Cell Rep 2025; 44:115439. [PMID: 40208794 PMCID: PMC12080591 DOI: 10.1016/j.celrep.2025.115439] [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: 05/14/2024] [Revised: 11/26/2024] [Accepted: 02/26/2025] [Indexed: 04/12/2025] Open
Abstract
Neurons dynamically regulate their proteome in response to sensory input, a key process underlying experience-dependent plasticity. We characterized the visual experience-dependent nascent proteome in mice within a brief, defined time window after stimulation using an optimized metabolic labeling approach. Visual experience induced cell-type-specific and age-dependent alterations in the nascent proteome, including proteostasis-related proteins. Emerin is the top activity-induced candidate plasticity protein. Activity-induced neuronal Emerin synthesis is rapid and transcription independent. Emerin broadly inhibits protein synthesis, decreasing translation regulators and synaptic proteins. Decreasing Emerin shifted the dendritic spine population from a predominantly mushroom morphology to filopodia and decreased network connectivity. Blocking visual experience-induced Emerin reduced visually evoked electrophysiological responses and impaired behaviorally assessed visual information processing. Our findings support a proteostatic model in which visual experience-induced Emerin provides a feedforward block on further protein synthesis, refining temporal control of activity-induced plasticity proteins and optimizing visual system function.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Graduate Program, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ruoxi Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel B McClatchy
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yuanhui Ma
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jolene Diedrich
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Manuel Sanchez-Alavez
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael Petrascheck
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John R Yates
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hollis T Cline
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
2
|
Song D, Gui F, Li G, Zhuang S, Sun J, Tan X, Hong C, Huang J. Neuritin improves cognitive impairments in APP/PS1 Alzheimer's disease mice model by mitigating neuronal ferroptosis via PI3K/Akt activation. Int J Biol Macromol 2025; 303:140662. [PMID: 39914536 DOI: 10.1016/j.ijbiomac.2025.140662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
The neurotrophic factor Neuritin is known to enhance cognitive capacity and to mitigate synaptic impairments in the APP/PS1 Alzheimer's disease (AD) mouse model, suggesting therapeutic potential for clinical treatment. However, the core molecular mechanisms remain elusive. Ferroptosis, a form of programmed cell death linked to iron dysregulation and oxidative stress, contributes to neurodegeneration in AD in part by accelerating amyloid-β deposition and neurofibrillary tangle formation. Here we examined if Neuritin can mitigate cognitive decline and neural degeneration in AD model mice by suppressing ferroptosis. Age-dependent cognitive decline was associated with Neuritin downregulation and increased ferroptosis in the hippocampus. Intracerebroventricular injection of exogenous Neuritin mitigated spatial and fear learning deficits as well as neural oxidative stress, apoptosis, and ferroptosis in the hippocampus without causing deleterious side effects. Neuritin injection also upregulated the activity of NAD+ kinase (NADK), the enzyme responsible for converting NAD to anti-ferroptotic NADPH, in the hippocampus of AD mice as well as in cultured hippocampal neurons. Reduced Neuritin expression in the hippocampus AD mice was associated with reduced phosphorylation (activation) of Akt (p-Akt), and Neuritin administration enhanced p-Akt expression in both HT22 cells and AD model mice. Conversely, blocking the PI3K/Akt pathway in HT22 cells reversed the Neuritin-induced increase in NADK activity and reduction in ferroptosis, indicating that Neuritin protects neurons from AD-induced damage by enhancing NADK activity through the PI3K/Akt pathway. Collectively, our results support Neuritin upregulation as a potential therapeutic strategy for early-phase AD.
Collapse
Affiliation(s)
- Dandan Song
- State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, School of Chemistry and Chemical Engineering, Shihezi University, Shihezi, Xinjiang 832003, PR China; Department of Preventive Medicine, School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, PR China
| | - Fei Gui
- Laboratory Animal Center, Hangzhou Normal University, Hangzhou, Zhejiang 311121, PR China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, PR China
| | - Guoxiang Li
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, PR China
| | - Shuai Zhuang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, PR China
| | - Jiawei Sun
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, PR China
| | - Xiaohua Tan
- Department of Preventive Medicine, School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, PR China.
| | - Chenglin Hong
- State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, School of Chemistry and Chemical Engineering, Shihezi University, Shihezi, Xinjiang 832003, PR China.
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, PR China.
| |
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
|
Hameed SS, Bodi NE, Miller RC, Sharma TP. Neuritin 1 Drives Therapeutic Preservation of Retinal Ganglion Cells in an Ex Vivo Human Glaucoma Model. J Ocul Pharmacol Ther 2024; 40:596-607. [PMID: 38995841 PMCID: PMC11698685 DOI: 10.1089/jop.2024.0041] [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: 02/26/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
Purpose: Glaucoma is a leading cause of irreversible blindness. Glaucomatous intraocular pressure (IOP) triggers deleterious effects, including gliosis, optic nerve (ON) axonal retraction, neurotrophic factor deprivation, inflammation, and other pathological events, leading to retinal ganglion cell (RGC) loss. Trophic factor impairment enhances RGC apoptosis susceptibility. Neuritin 1 (NRN1), a neurotrophic protein downstream of various neurotrophins, exhibited RGC protection and regeneration in axotomy models. We evaluated human recombinant NRN1's impact on human RGCs cultured in pressurized conditions within the ex vivo translaminar autonomous system to simulate glaucoma pathogenesis. Methods: Human glaucomatous and non-glaucomatous donor eyes were obtained from eye banks according to the Declaration of Helsinki. Initially, we evaluated NRN1and RGC marker expression in glaucoma and non-glaucomatous retina to determine the NRN1 level and its association with RGC loss. Further, we evaluated NRN1's therapeutic potential by treating pressurized human eyes at normal and high IOP for seven days. Retina, ON, and conditioned medium were analyzed for RGC survival (THY1, RBPMS), gliosis (GFAP), apoptosis (CASP3, CASP7), and extracellular matrix deposition (COLIV, FN) by qRT-PCR and western blotting. Paraphenylenediamine staining assessed ON axonal degeneration, whereas ex vivo electroretinogram assessed retinal activity. Results: Glaucomatous retinas exhibited significant reductions in both NRN1 (*p = 0.007, n = 5) and RGC marker expression (*p = 0.04, n = 5). NRN1 treatment reduced gliosis, extracellular matrix deposition, ON degeneration, and increased retinal activity in pressure-perfused eyes. Conclusions: Our study confirms that NRN1 enhances human RGC survival and improves retinal function in degenerative conditions, substantiating it as a promising candidate for rescuing human RGCs from degeneration.
Collapse
Affiliation(s)
- Shahna S. Hameed
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nicole E. Bodi
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ryan C. Miller
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Tasneem P. Sharma
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
5
|
Kuwako KI, Suzuki S. Diverse Roles of the LINC Complex in Cellular Function and Disease in the Nervous System. Int J Mol Sci 2024; 25:11525. [PMID: 39519078 PMCID: PMC11545860 DOI: 10.3390/ijms252111525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex, which spans the nuclear envelope, physically connects nuclear components to the cytoskeleton and plays a pivotal role in various cellular processes, including nuclear positioning, cell migration, and chromosomal configuration. Studies have revealed that the LINC complex is essential for different aspects of the nervous system, particularly during development. The significance of the LINC complex in neural lineage cells is further corroborated by the fact that mutations in genes associated with the LINC complex have been implicated in several neurological diseases, including neurodegenerative and psychiatric disorders. In this review, we aimed to summarize the expanding knowledge of LINC complex-related neuronal functions and associated neurological diseases.
Collapse
Affiliation(s)
- Ken-ichiro Kuwako
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi 693-8501, Shimane, Japan
| | | |
Collapse
|
6
|
Almodóvar-Payá C, Guardiola-Ripoll M, Giralt-López M, Oscoz-Irurozqui M, Canales-Rodríguez EJ, Madre M, Soler-Vidal J, Ramiro N, Callado LF, Arias B, Gallego C, Pomarol-Clotet E, Fatjó-Vilas M. NRN1 epistasis with BDNF and CACNA1C: mediation effects on symptom severity through neuroanatomical changes in schizophrenia. Brain Struct Funct 2024; 229:1299-1315. [PMID: 38720004 PMCID: PMC11147852 DOI: 10.1007/s00429-024-02793-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/19/2024] [Indexed: 06/05/2024]
Abstract
The expression of Neuritin-1 (NRN1), a neurotrophic factor crucial for neurodevelopment and synaptic plasticity, is enhanced by the Brain Derived Neurotrophic Factor (BDNF). Although the receptor of NRN1 remains unclear, it is suggested that NRN1's activation of the insulin receptor (IR) pathway promotes the transcription of the calcium voltage-gated channel subunit alpha1 C (CACNA1C). These three genes have been independently associated with schizophrenia (SZ) risk, symptomatology, and brain differences. However, research on how they synergistically modulate these phenotypes is scarce. We aimed to study whether the genetic epistasis between these genes affects the risk and clinical presentation of the disorder via its effect on brain structure. First, we tested the epistatic effect of NRN1 and BDNF or CACNA1C on (i) the risk for SZ, (ii) clinical symptoms severity and functionality (onset, PANSS, CGI and GAF), and (iii) brain cortical structure (thickness, surface area and volume measures estimated using FreeSurfer) in a sample of 86 SZ patients and 89 healthy subjects. Second, we explored whether those brain clusters influenced by epistatic effects mediate the clinical profiles. Although we did not find a direct epistatic impact on the risk, our data unveiled significant effects on the disorder's clinical presentation. Specifically, the NRN1-rs10484320 x BDNF-rs6265 interplay influenced PANSS general psychopathology, and the NRN1-rs4960155 x CACNA1C-rs1006737 interaction affected GAF scores. Moreover, several interactions between NRN1 SNPs and BDNF-rs6265 significantly influenced the surface area and cortical volume of the frontal, parietal, and temporal brain regions within patients. The NRN1-rs10484320 x BDNF-rs6265 epistasis in the left lateral orbitofrontal cortex fully mediated the effect on PANSS general psychopathology. Our study not only adds clinical significance to the well-described molecular relationship between NRN1 and BDNF but also underscores the utility of deconstructing SZ into biologically validated brain-imaging markers to explore their mediation role in the path from genetics to complex clinical manifestation.
Collapse
Affiliation(s)
- Carmen Almodóvar-Payá
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Guardiola-Ripoll
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- CIBERER (Biomedical Research Network in Rare Diseases), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Giralt-López
- Department of Child and Adolescent Psychiatry, Germans Trias i Pujol University Hospital (HUGTP), Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Faculty of Medicine, Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Maitane Oscoz-Irurozqui
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- Red de Salud Mental de Gipuzkoa, Osakidetza-Basque Health Service, Gipuzkoa, Spain
| | - Erick Jorge Canales-Rodríguez
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, Madrid, Spain
- Signal Processing Laboratory (LTS5), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Mercè Madre
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- Mental Health, IR SANT PAU, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Joan Soler-Vidal
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, Madrid, Spain
- Hospital Benito Menni, Germanes Hospitalàries, Sant Boi de Llobregat, Barcelona, Spain
| | - Núria Ramiro
- Hospital San Rafael, Germanes Hospitalàries, Barcelona, Spain
| | - Luis F Callado
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, Madrid, Spain
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Bizkaia, Spain
- BioBizkaia Health Research Institute, Bizkaia, Spain
| | - Bárbara Arias
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Carme Gallego
- Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Fatjó-Vilas
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain.
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
7
|
Devitt L, Westphal D, Pieger K, Schneider N, Bosserhoff AK, Kuphal S. NRN1 interacts with Notch to increase oncogenic STAT3 signaling in melanoma. Cell Commun Signal 2024; 22:256. [PMID: 38705997 PMCID: PMC11071257 DOI: 10.1186/s12964-024-01632-8] [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: 02/29/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Melanoma is a highly heterogeneous cancer, in which frequent changes in activation of signaling pathways lead to a high adaptability to ever changing tumor microenvironments. The elucidation of cancer specific signaling pathways is of great importance, as demonstrated by the inhibitor of the common BrafV600E mutation PLX4032 in melanoma treatment. We therefore investigated signaling pathways that were influenced by neurotrophin NRN1, which has been shown to be upregulated in melanoma. METHODS Using a cell culture model system with an NRN1 overexpression, we investigated the influence of NRN1 on melanoma cells' functionality and signaling. We employed real time cell analysis and spheroid formation assays, while for investigation of molecular mechanisms we used a kinase phosphorylation kit as well as promotor activity analysis followed by mRNA and protein analysis. RESULTS We revealed that NRN1 interacts directly with the cleaved intracellular domain (NICD) of Notch1 and Notch3, causing a potential retention of NICD in the cytoplasm and thereby reducing the expression of its direct downstream target Hes1. This leads to decreased sequestration of JAK and STAT3 in a Hes1-driven phosphorylation complex. Consequently, our data shows less phosphorylation of STAT3 while presenting an accumulation of total protein levels of STAT3 in association with NRN1 overexpression. The potential of the STAT3 signaling pathway to act in both a tumor suppressive and oncogenic manner led us to investigate specific downstream targets - namely Vegf A, Mdr1, cMet - which were found to be upregulated under oncogenic levels of NRN1. CONCLUSIONS In summary, we were able to show that NRN1 links oncogenic signaling events between Notch and STAT3 in melanoma. We also suggest that in future research more attention should be payed to cellular regulation of signaling molecules outside of the classically known phosphorylation events.
Collapse
Affiliation(s)
- Lucia Devitt
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstrasse 17, Erlangen, 91054, Germany
| | - Dana Westphal
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus at TU Dresden, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Katharina Pieger
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstrasse 17, Erlangen, 91054, Germany
| | - Nadja Schneider
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstrasse 17, Erlangen, 91054, Germany
| | - Anja Katrin Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstrasse 17, Erlangen, 91054, Germany
| | - Silke Kuphal
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstrasse 17, Erlangen, 91054, Germany.
| |
Collapse
|
8
|
Papadogianni G, Ravens I, Hassan A, Flatley A, Feederle R, Bernhardt G, Georgiev H. Establishment and Functional Characterization of Murine Monoclonal Antibodies Recognizing Neuritin. Antibodies (Basel) 2023; 12:antib12020028. [PMID: 37092449 PMCID: PMC10123642 DOI: 10.3390/antib12020028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/17/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Neuritin represents a neurotrophic factor that is not only important in neuronal development and plasticity but also impacts endothelial angiogenesis, cell migration, tumor growth and the production of antibodies by B cells. We established monoclonal mouse anti-mouse neuritin antibodies by immunizing knock-out mice with two different neuritin-derived peptides. Because neuritin is well conserved between species, these new monoclonal antibodies recognize the neuritin of a wide variety of species, including human. Moreover, they not only recognize specifically surface-bound neuritin expressed by murine follicular regulatory T cells but also the block binding of recombinant neuritin to germinal center B cells. This suggests that these newly generated tools will be of great use in studying neuritin expression and function.
Collapse
Affiliation(s)
| | - Inga Ravens
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Ahmed Hassan
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Andrew Flatley
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Hristo Georgiev
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
9
|
Bhat VD, Jayaraj J, Babu K. RNA and neuronal function: the importance of post-transcriptional regulation. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac011. [PMID: 38596700 PMCID: PMC10913846 DOI: 10.1093/oons/kvac011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/03/2022] [Accepted: 05/28/2022] [Indexed: 04/11/2024]
Abstract
The brain represents an organ with a particularly high diversity of genes that undergo post-transcriptional gene regulation through multiple mechanisms that affect RNA metabolism and, consequently, brain function. This vast regulatory process in the brain allows for a tight spatiotemporal control over protein expression, a necessary factor due to the unique morphologies of neurons. The numerous mechanisms of post-transcriptional regulation or translational control of gene expression in the brain include alternative splicing, RNA editing, mRNA stability and transport. A large number of trans-elements such as RNA-binding proteins and micro RNAs bind to specific cis-elements on transcripts to dictate the fate of mRNAs including its stability, localization, activation and degradation. Several trans-elements are exemplary regulators of translation, employing multiple cofactors and regulatory machinery so as to influence mRNA fate. Networks of regulatory trans-elements exert control over key neuronal processes such as neurogenesis, synaptic transmission and plasticity. Perturbations in these networks may directly or indirectly cause neuropsychiatric and neurodegenerative disorders. We will be reviewing multiple mechanisms of gene regulation by trans-elements occurring specifically in neurons.
Collapse
Affiliation(s)
- Vandita D Bhat
- Centre for Neuroscience, Indian Institute of Science, CV Raman Road, Bangalore 560012, Karnataka, India
| | - Jagannath Jayaraj
- Centre for Neuroscience, Indian Institute of Science, CV Raman Road, Bangalore 560012, Karnataka, India
| | - Kavita Babu
- Centre for Neuroscience, Indian Institute of Science, CV Raman Road, Bangalore 560012, Karnataka, India
| |
Collapse
|
10
|
Almodóvar-Payá C, Guardiola-Ripoll M, Giralt-López M, Gallego C, Salgado-Pineda P, Miret S, Salvador R, Muñoz MJ, Lázaro L, Guerrero-Pedraza A, Parellada M, Carrión MI, Cuesta MJ, Maristany T, Sarró S, Fañanás L, Callado LF, Arias B, Pomarol-Clotet E, Fatjó-Vilas M. NRN1 Gene as a Potential Marker of Early-Onset Schizophrenia: Evidence from Genetic and Neuroimaging Approaches. Int J Mol Sci 2022; 23:ijms23137456. [PMID: 35806464 PMCID: PMC9267632 DOI: 10.3390/ijms23137456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/10/2022] Open
Abstract
Included in the neurotrophins family, the Neuritin 1 gene (NRN1) has emerged as an attractive candidate gene for schizophrenia (SZ) since it has been associated with the risk for the disorder and general cognitive performance. In this work, we aimed to further investigate the association of NRN1 with SZ by exploring its role on age at onset and its brain activity correlates. First, we developed two genetic association analyses using a family-based sample (80 early-onset (EO) trios (offspring onset ≤ 18 years) and 71 adult-onset (AO) trios) and an independent case–control sample (120 healthy subjects (HS), 87 EO and 138 AO patients). Second, we explored the effect of NRN1 on brain activity during a working memory task (N-back task; 39 HS, 39 EO and 39 AO; matched by age, sex and estimated IQ). Different haplotypes encompassing the same three Single Nucleotide Polymorphisms(SNPs, rs3763180–rs10484320–rs4960155) were associated with EO in the two samples (GCT, TCC and GTT). Besides, the GTT haplotype was associated with worse N-back task performance in EO and was linked to an inefficient dorsolateral prefrontal cortex activity in subjects with EO compared to HS. Our results show convergent evidence on the NRN1 association with EO both from genetic and neuroimaging approaches, highlighting the role of neurotrophins in the pathophysiology of SZ.
Collapse
Affiliation(s)
- Carmen Almodóvar-Payá
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - Maria Guardiola-Ripoll
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - Maria Giralt-López
- Departament de Psiquiatria, Hospital Universitari Germans Trias i Pujol (HUGTP), 08916 Badalona, Barcelona, Spain;
- Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Barcelona, Spain
| | - Carme Gallego
- Department of Cell Biology, Molecular Biology Institute of Barcelona (IBMB-CSIC), 08028 Barcelona, Barcelona, Spain;
| | - Pilar Salgado-Pineda
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - Salvador Miret
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Centre de Salut Mental d’Adults de Lleida, Servei de Psiquiatria, Salut Mental i Addiccions, Hospital Universitari Santa Maria de Lleida, 25198 Lleida, Lleida, Spain
- Institut de Recerca Biomèdica (IRB), 25198 Lleida, Lleida, Spain
| | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - María J. Muñoz
- Complex Assistencial en Salut Mental Benito Menni, 08830 Sant Boi de Llobregat, Barcelona, Spain;
| | - Luisa Lázaro
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de Barcelona, 08036 Barcelona, Barcelona, Spain
- Departament de Medicina, Universitat de Barcelona (UB), 08036 Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Barcelona, Spain
| | - Amalia Guerrero-Pedraza
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Complex Assistencial en Salut Mental Benito Menni, 08830 Sant Boi de Llobregat, Barcelona, Spain;
| | - Mara Parellada
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Servicio de Psiquiatría del Niño y del Adolescente, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Gregorio Marañón (IiSGM), 28007 Madrid, Madrid, Spain
- Departamento de Psiquiatría, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Madrid, Spain
| | | | - Manuel J. Cuesta
- Servicio de Psiquiatría, Hospital Universitario de Navarra, 31008 Pamplona, Navarra, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Navarra, Spain
| | - Teresa Maristany
- Departament de Diagnòstic per la Imatge, Hospital Sant Joan de Déu Fundació de Recerca, 08950 Esplugues de Llobregat, Barcelona, Spain;
| | - Salvador Sarró
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - Lourdes Fañanás
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Departament de Biologia Evolutiva, Ecología i Ciències Ambientals, Universitat de Barcelona (UB), 08028 Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Barcelona, Spain
| | - Luis F. Callado
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Bizkaia, Spain
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Bárbara Arias
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Departament de Biologia Evolutiva, Ecología i Ciències Ambientals, Universitat de Barcelona (UB), 08028 Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Barcelona, Spain
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Correspondence: (E.P.-C.); (M.F.-V.)
| | - Mar Fatjó-Vilas
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Departament de Biologia Evolutiva, Ecología i Ciències Ambientals, Universitat de Barcelona (UB), 08028 Barcelona, Barcelona, Spain
- Correspondence: (E.P.-C.); (M.F.-V.)
| |
Collapse
|
11
|
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
|
12
|
Utsunomiya S, Kishi Y, Tsuboi M, Kawaguchi D, Gotoh Y, Abe M, Sakimura K, Maeda K, Takemoto H. Ezh1 regulates expression of Cpg15/Neuritin in mouse cortical neurons. Drug Discov Ther 2021; 15:55-65. [PMID: 33678755 DOI: 10.5582/ddt.2021.01017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immature neurons undergo morphological and physiological maturation in order to establish neuronal networks. During neuronal maturation, a large number of genes change their transcriptional levels, and these changes may be mediated by chromatin modifiers. In this study, we found that the level of Ezh1, a component of Polycomb repressive complex 2 (PRC2), increases during neuronal maturation in mouse neocortical culture. In addition, conditional knockout of Ezh1 in post-mitotic excitatory neurons leads to downregulation of a set of genes related to neuronal maturation. Moreover, the locus encoding Cpg15/Neuritin (Nrn1), which is regulated by neuronal activity and implicated in stabilization and maturation of excitatory synapses, is a direct target of Ezh1 in cortical neurons. Together, these results suggest that elevated expression of Ezh1 contributes to maturation of cortical neurons.
Collapse
Affiliation(s)
- Shun Utsunomiya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan.,Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yusuke Kishi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Masafumi Tsuboi
- Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Daichi Kawaguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kazuma Maeda
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan.,Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Takemoto
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan.,Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
The why and how of sleep-dependent synaptic down-selection. Semin Cell Dev Biol 2021; 125:91-100. [PMID: 33712366 PMCID: PMC8426406 DOI: 10.1016/j.semcdb.2021.02.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/28/2021] [Accepted: 02/28/2021] [Indexed: 12/22/2022]
Abstract
Sleep requires that we disconnect from the environment, losing the ability to promptly respond to stimuli. There must be at least one essential function that justifies why we take this risk every day, and that function must depend on the brain being offline. We have proposed that this function is to renormalize synaptic weights after learning has led to a net increase in synaptic strength in many brain circuits. Without this renormalization, synaptic activity would become energetically too expensive and saturation would prevent new learning. There is converging evidence from molecular, electrophysiological, and ultrastructural experiments showing a net increase in synaptic strength after the major wake phase, and a net decline after sleep. The evidence also suggests that sleep-dependent renormalization is a smart process of synaptic down-selection, comprehensive and yet specific, which could explain the many beneficial effects of sleep on cognition. Recently, a key molecular mechanism that allows broad synaptic weakening during sleep was identified. Other mechanisms still being investigated should eventually explain how sleep can weaken most synapses but afford protection to some, including those directly activated by learning. That synaptic down-selection takes place during sleep is by now established; why it should take place during sleep has a plausible explanation; how it happens is still work in progress.
Collapse
|
14
|
Neuritin-overexpressing transgenic mice demonstrate enhanced neuroregeneration capacity and improved spatial learning and memory recovery after ischemia-reperfusion injury. Aging (Albany NY) 2020; 13:2681-2699. [PMID: 33323541 PMCID: PMC7880330 DOI: 10.18632/aging.202318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/01/2020] [Indexed: 02/01/2023]
Abstract
Acute ischemia-reperfusion (IR)-induced brain injury is further exacerbated by a series of slower secondary pathogenic events, including delayed apoptosis due to neurotrophic factor deficiency. Neuritin, a neurotrophic factor regulating nervous system development and plasticity, is a potential therapeutic target for treatment of IR injury. In this study, Neuritin-overexpressing transgenic (Tg) mice were produced by pronuclear injection and offspring with high overexpression used to generate a line with stable inheritance for testing the neuroprotective capacity of Neuritin against transient global ischemia (TGI). Compared to wild-type mice, transgenic mice demonstrated reduced degradation of the DNA repair factor poly [ADP-ribose] polymerase 1 (PARP 1) in the hippocampus, indicating decreased hippocampal apoptosis rate, and a greater number of surviving hippocampal neurons during the first week post-TGI. In addition, Tg mice showed increased expression of the regeneration markers NF-200, synaptophysin, and GAP-43, and improved recovery of spatial learning and memory. Our findings exhibited that the window of opportunity of neural recovery in Neuritin transgenic mice group had a tendency to move ahead after TGI, which indicated that Neuritin can be used as a potential new therapeutic strategy for improving the outcome of cerebral ischemia injury.
Collapse
|
15
|
Yu L, Tasaki S, Schneider JA, Arfanakis K, Duong DM, Wingo AP, Wingo TS, Kearns N, Thatcher GRJ, Seyfried NT, Levey AI, De Jager PL, Bennett DA. Cortical Proteins Associated With Cognitive Resilience in Community-Dwelling Older Persons. JAMA Psychiatry 2020; 77:1172-1180. [PMID: 32609320 PMCID: PMC7330835 DOI: 10.1001/jamapsychiatry.2020.1807] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
Abstract
Importance Identifying genes and proteins for cognitive resilience (ie, targets that may be associated with slowing or preventing cognitive decline regardless of the presence, number, or combination of common neuropathologic conditions) provides a complementary approach to developing novel therapeutics for the treatment and prevention of Alzheimer disease and related dementias. Objective To identify proteins associated with cognitive resilience via a proteome-wide association study of the human dorsolateral prefrontal cortex. Design, Setting, and Participants This study used data from 391 community-dwelling older persons who participated in the Religious Orders Study and the Rush Memory and Aging Project. The Religious Orders Study began enrollment January 1, 1994, and the Rush Memory and Aging Project began enrollment September 1, 1997, and data were collected and analyzed through October 23, 2019. Exposures Participants had undergone annual detailed clinical examinations, postmortem evaluations, and tandem mass tag proteomics analyses. Main Outcomes and Measures The outcome of cognitive resilience was defined as a longitudinal change in cognition over time after controlling for common age-related neuropathologic indices, including Alzheimer disease, Lewy bodies, transactive response DNA-binding protein 43, hippocampal sclerosis, infarcts, and vessel diseases. More than 8000 high abundance proteins were quantified from frozen dorsolateral prefrontal cortex tissue using tandem mass tag and liquid chromatography-mass spectrometry. Results There were 391 participants (273 women); their mean (SD) age was 79.7 (6.7) years at baseline and 89.2 (6.5) years at death. Eight cortical proteins were identified in association with cognitive resilience: a higher level of NRN1 (estimate, 0.140; SE, 0.024; P = 7.35 × 10-9), ACTN4 (estimate, 0.321; SE, 0.065; P = 9.94 × 10-7), EPHX4 (estimate, 0.198; SE, 0.042; P = 2.13 × 10-6), RPH3A (estimate, 0.148; SE, 0.031; P = 2.58 × 10-6), SGTB (estimate, 0.211; SE, 0.045; P = 3.28 × 10-6), CPLX1 (estimate, 0.136; SE, 0.029; P = 4.06 × 10-6), and SH3GL1 (estimate, 0.179; SE, 0.039; P = 4.21 × 10-6) and a lower level of UBA1 (estimate, -0.366; SE, 0.076; P = 1.43 × 10-6) were associated with greater resilience. Conclusions and Relevance These protein signals may represent novel targets for the maintenance of cognition in old age.
Collapse
Affiliation(s)
- Lei Yu
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Shinya Tasaki
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
- Department of Pathology, Rush University Medical Center, Chicago, Illinois
| | - Konstantinos Arfanakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, Illinois
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago
| | - Duc M. Duong
- Department of Biochemistry, Emory University, Atlanta, Georgia
| | - Aliza P. Wingo
- Division of Mental Health, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
- Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia
| | - Thomas S. Wingo
- Department of Neurology, Emory University, Atlanta, Georgia
- Department of Human Genetics, Emory University, Atlanta, Georgia
| | - Nicola Kearns
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Gregory R. J. Thatcher
- Department of Pharmaceutical Sciences, University of Illinois College of Pharmacy, Chicago
| | | | - Allan I. Levey
- Department of Neurology, Emory University, Atlanta, Georgia
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, New York
- Cell Circuits Program, Broad Institute, Cambridge, Massachusetts
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
16
|
CPG15/Neuritin Mimics Experience in Selecting Excitatory Synapses for Stabilization by Facilitating PSD95 Recruitment. Cell Rep 2020; 28:1584-1595.e5. [PMID: 31390571 PMCID: PMC6740334 DOI: 10.1016/j.celrep.2019.07.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/11/2019] [Accepted: 07/02/2019] [Indexed: 11/24/2022] Open
Abstract
A key feature of brain plasticity is the experience-dependent selection of optimal connections· implemented by a set of activity-regulated genes that dynamically adjust synapse strength and number. The activity-regulated gene cpg15/neuritin has been previously implicated in stabilization and maturation of excitatory synapses. Here· we combine two-photon microscopy with genetic and sensory manipulations to dissect excitatory synapse formation in vivo and examine the role of activity and CPG15 in dendritic spine formation, PSD95 recruitment, and synapse stabilization. We find that neither visual experience nor CPG15 is required for spine formation. However, PSD95 recruitment to nascent spines and their subsequent stabilization requires both. Further, cell-autonomous CPG15 expression is sufficient to replace experience in facilitating PSD95 recruitment and spine stabilization. CPG15 directly interacts with α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors on immature dendritic spines, suggesting a signaling mode for this small extracellular molecule acting as an experience-dependent “selector” for spine stabilization and synapse maturation. Experience plays a key role in formation and continuous optimization of brain circuits. Subramanian et al. show that the molecule CPG15/neuritin can replace experience in selecting which nascent contacts between neurons are retained, facilitating the recruitment of proteins that promote synapse maturation and stabilization.
Collapse
|
17
|
Runge K, Cardoso C, de Chevigny A. Dendritic Spine Plasticity: Function and Mechanisms. Front Synaptic Neurosci 2020. [DOI: 10.3389/fnsyn.2020.00036
expr 823669561 + 872784217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
|
18
|
Runge K, Cardoso C, de Chevigny A. Dendritic Spine Plasticity: Function and Mechanisms. Front Synaptic Neurosci 2020; 12:36. [PMID: 32982715 PMCID: PMC7484486 DOI: 10.3389/fnsyn.2020.00036] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic spines are small protrusions studding neuronal dendrites, first described in 1888 by Ramón y Cajal using his famous Golgi stainings. Around 50 years later the advance of electron microscopy (EM) confirmed Cajal's intuition that spines constitute the postsynaptic site of most excitatory synapses in the mammalian brain. The finding that spine density decreases between young and adult ages in fixed tissues suggested that spines are dynamic. It is only a decade ago that two-photon microscopy (TPM) has unambiguously proven the dynamic nature of spines, through the repeated imaging of single spines in live animals. Spine dynamics comprise formation, disappearance, and stabilization of spines and are modulated by neuronal activity and developmental age. Here, we review several emerging concepts in the field that start to answer the following key questions: What are the external signals triggering spine dynamics and the molecular mechanisms involved? What is, in return, the role of spine dynamics in circuit-rewiring, learning, and neuropsychiatric disorders?
Collapse
Affiliation(s)
- Karen Runge
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| | - Carlos Cardoso
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| | - Antoine de Chevigny
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| |
Collapse
|
19
|
MEF2C and HDAC5 regulate Egr1 and Arc genes to increase dendritic spine density and complexity in early enriched environment. Neuronal Signal 2020; 4:NS20190147. [PMID: 32714604 PMCID: PMC7378308 DOI: 10.1042/ns20190147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 01/16/2023] Open
Abstract
We investigated the effects of environmental enrichment during critical period of early postnatal life and how it interplays with the epigenome to affect experience-dependent visual cortical plasticity. Mice raised in an EE from birth to during CP have increased spine density and dendritic complexity in the visual cortex. EE upregulates synaptic plasticity genes, Arc and Egr1, and a transcription factor MEF2C. We also observed an increase in MEF2C binding to the promoters of Arc and Egr1. In addition, pups raised in EE show a reduction in HDAC5 and its binding to promoters of Mef2c, Arc and Egr1 genes. With an overexpression of Mef2c, neurite outgrowth increased in complexity. Our results suggest a possible underlying molecular mechanism of EE, acting through MEF2C and HDAC5, which drive Arc and Egr1. This could lead to the observed increased dendritic spine density and complexity induced by early EE.
Collapse
|
20
|
Decreased cpg15 augments oxidative stress in sleep deprived mouse brain. Biochem Biophys Res Commun 2019; 522:749-756. [PMID: 31787230 DOI: 10.1016/j.bbrc.2019.11.132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 11/20/2019] [Indexed: 11/21/2022]
Abstract
Sleep deprivation (SD) has detrimental effects on the physiological function of the brain. However, the underlying mechanism remains elusive. In the present study, we investigated the expression of candidate plasticity-related gene 15 (cpg15), a neurotrophic gene, and its potential role in SD using a REM-SD mouse model. Immunofluorescent and Western blot analysis revealed that the expression of cpg15 protein decreased in the hippocampus, ventral group of the dorsal thalamus (VENT), and somatosensory area of cerebral cortex (SSP) after 24-72 h of REM-SD, and the oxidative stress in these brain regions was increased in parallel, as indicated by the ratio of glutathione (GSH) to its oxidative product (GSSG). Over-expression of cpg15 in thalamus, hippocampus, and cerebral cortex mediated by AAV reduced the oxidative stress in these regions, indicating that the decrease of cpg15 might be a cause that augments oxidative stress in the sleep deprived mouse brain. Collectively, the results imply that cpg15 may play a protective function in the SD-subjected mouse brain via an anti-oxidative function. To our knowledge, this is the first time to provide evidences in the role of cpg15 against SD-induced oxidative stress in the brain.
Collapse
|
21
|
Wang T, Cheng Y, Han H, Liu J, Tian B, Liu X. miR-194 Accelerates Apoptosis of Aβ 1⁻42-Transduced Hippocampal Neurons by Inhibiting Nrn1 and Decreasing PI3K/Akt Signaling Pathway Activity. Genes (Basel) 2019; 10:genes10040313. [PMID: 31010100 PMCID: PMC6523401 DOI: 10.3390/genes10040313] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022] Open
Abstract
This article explores the mechanism of miR-194 on the proliferation and apoptosis of Aβ1–42-transduced hippocampal neurons. Aβ1–42-transduced hippocampal neuron model was established by inducing hippocampal neurons with Aβ1–42. MTT assay and flow cytometry were used to detect the viability and apoptosis of hippocampal neurons, respectively. qRT-PCR was used to detect changes in miR-194 and Nrn1 expression after Aβ1–42 induction. Aβ1–42-transduced hippocampal neurons were transfected with miR-194 mimics and/or Nrn1 overexpression vectors. Their viability and neurite length were detected by MTT assay and immunofluorescence, respectively. Western blot was used to detect protein expression. Aβ1–42 inhibited Aβ1–42-transduced hippocampal neuron activity and promoted their apoptosis in a dose-dependent manner. miR-194 was upregulated and Nrn1 was downregulated in Aβ1–42-transduced hippocampal neurons (p < 0.05). Compared with the model group, Aβ1–42-transduced hippocampal neurons of the miR-194 mimic group had much lower activity, average longest neurite length, Nrn1, p-AkT, and Bcl-2 protein expression and had much higher Bax, Caspase-3, and Cleaved Caspase-3 protein expression. Compared with the model group, Aβ1–42-transduced hippocampal neurons of the LV-Nrn1 group had much higher activity, average longest neurite length, Nrn1, p-AkT, and Bcl-2 protein expression and had much lower Bax, Caspase-3, and Cleaved Caspase-3 protein expression. Nrn1 is a target gene of miR-194. miR-194 inhibited apoptosis of Aβ1–42-transduced hippocampal neurons by inhibiting Nrn1 and decreasing PI3K/AkT signaling pathway activity.
Collapse
Affiliation(s)
- Tingting Wang
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Yaling Cheng
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Haibin Han
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Jie Liu
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Bo Tian
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Xiaocui Liu
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| |
Collapse
|
22
|
Wang N, Wei Y, Zhang W, Li X, Zhu J, Shan L, Liu C, Yuan W, Huang J. Production of polyclonal antibody against human Neuritin and its application of immunodetection. Prep Biochem Biotechnol 2019; 49:209-214. [PMID: 30822252 DOI: 10.1080/10826068.2018.1476877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To date, a commercial antibody to human Neuritin for immunoprecipitation is still limited. In this study, we aimed to develop a specific antibody for further research on the potential function of Neuritin. METHODS AND RESULTS By epitope prediction of recombinant human Neuritin, the active fragment of human Neuritin that could be used as an excellent immunogen. Soluble His-tagged Neuritin was expressed and purified from Pichia pastoris. Polyclonal antibody against Neuritin was obtained by immunizing Sprague-Dawley rats with purified recombinant human Neuritin. Affinity-purified polyclonal antibody against Neuritin was characterized with indirect enzyme-linked immunosorbent assay, immunoblotting, immunoprecipitation, and immunofluorescence. The results demonstrated that the polyclonal antibody against Neuritin had been prepared successfully. The prepared antibody bound to both exogenous and endogenous Neuritin. Importantly, the anti-Neuritin polyclonal antibody could be used in immunoprecipitation assays. CONCLUSIONS The prepared polyclonal antibody could be used in immunoprecipitation and provide researchers with a useful tool for further investigating the function and mechanism of Neuritin.
Collapse
Affiliation(s)
- Na Wang
- a The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry , Shihezi University School of Medicine , Shihezi , Xinjiang , China
| | - Yu Wei
- b The First Affiliated Hospital of the Medical College, Shihezi University , Shihezi , Xinjiang , China
| | - Wen Zhang
- a The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry , Shihezi University School of Medicine , Shihezi , Xinjiang , China
| | - Xingyi Li
- c Livzon Pharmaceutical Group Inc., Clinical Research Center , Guangdong , China
| | - Jingling Zhu
- a The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry , Shihezi University School of Medicine , Shihezi , Xinjiang , China
| | - Liya Shan
- a The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry , Shihezi University School of Medicine , Shihezi , Xinjiang , China
| | - Chunyan Liu
- a The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry , Shihezi University School of Medicine , Shihezi , Xinjiang , China
| | - Wumei Yuan
- a The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry , Shihezi University School of Medicine , Shihezi , Xinjiang , China
| | - Jin Huang
- a The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry , Shihezi University School of Medicine , Shihezi , Xinjiang , China
| |
Collapse
|
23
|
Identification of rare nonsynonymous variants in SYNE1/CPG2 in bipolar affective disorder. Psychiatr Genet 2018; 27:81-88. [PMID: 28178086 DOI: 10.1097/ypg.0000000000000166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Bipolar affective disorder (BPD) is a severe mood disorder with a prevalence of ∼1.5% in the population. The pathogenesis of BPD is poorly understood; however, a strong heritable component has been identified. Previous genome-wide association studies have indicated a region on 6q25, coding for the SYNE1 gene, which increases disease susceptibility. SYNE1 encodes the synaptic nuclear envelope protein-1, nesprin-1. A brain-specific splice variant of SYNE1, CPG2 encoding candidate plasticity gene 2, has been identified. The intronic single-nucleotide polymorphism with the strongest genome-wide significant association in BPD, rs9371601, is present in both SYNE1 and CPG2. METHODS We screened 937 BPD samples for genetic variation in SYNE1 exons 14-33, which covers the CPG2 region, using high-resolution melt analysis. In addition, we screened two regions of increased transcriptional activity, one of them proposed to be the CPG2 promoter region. RESULTS AND CONCLUSION We identified six nonsynonymous and six synonymous variants. We genotyped three rare nonsynonymous variants, rs374866393, rs148346599 and rs200629713, in a total of 1099 BPD samples and 1056 controls. Burden analysis of these rare variants did not show a significant association with BPD. However, nine patients are compound heterozygotes for variants in SYNE1/CPG2, suggesting that rare coding variants may contribute significantly towards the complex genetic architecture underlying BPD. Imputation analysis in our own whole-genome sequencing sample of 99 BPD individuals identified an additional eight risk variants in the CPG2 region of SYNE1.
Collapse
|
24
|
Khalil R, Contreras-Ramirez V, Levitt JB. Postnatal refinement of interareal feedforward projections in ferret visual cortex. Brain Struct Funct 2018; 223:2303-2322. [PMID: 29476239 DOI: 10.1007/s00429-018-1632-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 02/17/2018] [Indexed: 12/27/2022]
Abstract
We studied the postnatal refinement of feedforward (FF) projections from ferret V1 to multiple cortical targets during the period around eye opening. Our goal was to establish (a) whether the developmental refinement of FF projections parallels that of feedback (FB) cortical circuits, and (b) whether FF pathways from V1 to different target areas refine with a similar rate. We injected the tracer CTb into V1 of juvenile ferrets, and visualized the pattern of labeled axon terminals in extrastriate cortex. Bouton density of FF projections to target areas 18, 19, and 21 declined steadily from 4 to 8 weeks postnatal. However, in area Ssy this decline was delayed somewhat, not occurring until after 6 weeks. During this postnatal period, mean interbouton intervals along individual FF axons to all visual areas increased, and we observed a concomitant moderate decrease in axon density in areas 18, 21, and Ssy. These data suggest that FF circuits linking V1 to its main extrastriate targets remodel largely synchronously in the weeks following eye opening, that FF and FB cortical circuits share a broadly similar developmental timecourse, and that postnatal visual experience is critical for the refinement of both FF and FB cortical circuits.
Collapse
Affiliation(s)
- Reem Khalil
- Biology, Chemistry, and Environmental Sciences Department, American University of Sharjah, Sharjah, UAE.,Department of Biology MR526, City College of New York, 160 Convent Avenue, New York, NY, 10031, USA.,Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA
| | | | - Jonathan B Levitt
- Department of Biology MR526, City College of New York, 160 Convent Avenue, New York, NY, 10031, USA. .,Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA.
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
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]
|
28
|
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
|
29
|
Steinmetz CC, Tatavarty V, Sugino K, Shima Y, Joseph A, Lin H, Rutlin M, Lambo M, Hempel CM, Okaty BW, Paradis S, Nelson SB, Turrigiano GG. Upregulation of μ3A Drives Homeostatic Plasticity by Rerouting AMPAR into the Recycling Endosomal Pathway. Cell Rep 2016; 16:2711-2722. [PMID: 27568566 DOI: 10.1016/j.celrep.2016.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 07/15/2016] [Accepted: 08/01/2016] [Indexed: 01/06/2023] Open
Abstract
Synaptic scaling is a form of homeostatic plasticity driven by transcription-dependent changes in AMPA-type glutamate receptor (AMPAR) trafficking. To uncover the pathways involved, we performed a cell-type-specific screen for transcripts persistently altered during scaling, which identified the μ subunit (μ3A) of the adaptor protein complex AP-3A. Synaptic scaling increased μ3A (but not other AP-3 subunits) in pyramidal neurons and redistributed dendritic μ3A and AMPAR to recycling endosomes (REs). Knockdown of μ3A prevented synaptic scaling and this redistribution, while overexpression (OE) of full-length μ3A or a truncated μ3A that cannot interact with the AP-3A complex was sufficient to drive AMPAR to REs. Finally, OE of μ3A acted synergistically with GRIP1 to recruit AMPAR to the dendritic membrane. These data suggest that excess μ3A acts independently of the AP-3A complex to reroute AMPAR to RE, generating a reservoir of receptors essential for the regulated recruitment to the synaptic membrane during scaling up.
Collapse
Affiliation(s)
- Celine C Steinmetz
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Vedakumar Tatavarty
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Ken Sugino
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Yasuyuki Shima
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Anne Joseph
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Heather Lin
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Michael Rutlin
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Mary Lambo
- Department of Brain and Cognitive Science, MIT, Cambridge, MA 02139, USA
| | - Chris M Hempel
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Benjamin W Okaty
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Suzanne Paradis
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Sacha B Nelson
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA.
| | - Gina G Turrigiano
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA.
| |
Collapse
|
30
|
Sun X, Lin Y. Npas4: Linking Neuronal Activity to Memory. Trends Neurosci 2016; 39:264-275. [PMID: 26987258 DOI: 10.1016/j.tins.2016.02.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/03/2016] [Accepted: 02/09/2016] [Indexed: 01/16/2023]
Abstract
Immediate-early genes (IEGs) are rapidly activated after sensory and behavioral experience and are believed to be crucial for converting experience into long-term memory. Neuronal PAS domain protein 4 (Npas4), a recently discovered IEG, has several characteristics that make it likely to be a particularly important molecular link between neuronal activity and memory: it is among the most rapidly induced IEGs, is expressed only in neurons, and is selectively induced by neuronal activity. By orchestrating distinct activity-dependent gene programs in different neuronal populations, Npas4 affects synaptic connections in excitatory and inhibitory neurons, neural circuit plasticity, and memory formation. It may also be involved in circuit homeostasis through negative feedback and psychiatric disorders. We summarize these findings and discuss their implications.
Collapse
Affiliation(s)
- Xiaochen Sun
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Molecular and Cellular Neuroscience Graduate Program, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Yingxi Lin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
31
|
Loebrich S, Rathje M, Hager E, Ataman B, Harmin DA, Greenberg ME, Nedivi E. Genomic mapping and cellular expression of human CPG2 transcripts in the SYNE1 gene. Mol Cell Neurosci 2015; 71:46-55. [PMID: 26704904 DOI: 10.1016/j.mcn.2015.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 01/13/2023] Open
Abstract
Bipolar disorder (BD) is a prevalent and severe mood disorder characterized by recurrent episodes of mania and depression. Both genetic and environmental factors have been implicated in BD etiology, but the biological underpinnings remain elusive. Recent genome-wide association studies (GWAS) for identifying genes conferring risk for schizophrenia, BD, and major depression, identified an association between single-nucleotide polymorphisms (SNPs) in the SYNE1 gene and increased risk of BD. SYNE1 has also been identified as a risk locus for multiple other neurological or neuromuscular genetic disorders. The BD associated SNPs map within the gene region homologous to part of rat Syne1 encompassing the brain specific transcripts encoding CPG2, a postsynaptic neuronal protein localized to excitatory synapses and an important regulator of glutamate receptor internalization. Here, we use RNA-seq, ChIP-seq and RACE to map the human SYNE1 transcriptome, focusing on the CPG2 locus. We validate several CPG2 transcripts, including ones not previously annotated in public databases, and identify and clone a full-length CPG2 cDNA expressed in human neocortex, hippocampus and striatum. Using lenti-viral gene knock down/replacement and surface receptor internalization assays, we demonstrate that human CPG2 protein localizes to dendritic spines in rat hippocampal neurons and is functionally equivalent to rat CPG2 in regulating glutamate receptor internalization. This study provides a valuable gene-mapping framework for relating multiple genetic disease loci in SYNE1 with their transcripts, and for evaluating the effects of missense SNPs identified by patient genome sequencing on neuronal function.
Collapse
Affiliation(s)
- Sven Loebrich
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mette Rathje
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Emily Hager
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bulent Ataman
- Department of Neurobiology, Harvard Medical School, Boston, MA 02114, USA
| | - David A Harmin
- Department of Neurobiology, Harvard Medical School, Boston, MA 02114, USA
| | | | - Elly Nedivi
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
32
|
Sachdeva R, Theisen CC, Ninan V, Twiss JL, Houlé JD. Exercise dependent increase in axon regeneration into peripheral nerve grafts by propriospinal but not sensory neurons after spinal cord injury is associated with modulation of regeneration-associated genes. Exp Neurol 2015; 276:72-82. [PMID: 26366525 DOI: 10.1016/j.expneurol.2015.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 07/28/2015] [Accepted: 09/08/2015] [Indexed: 01/04/2023]
Abstract
Insufficient regeneration of central nervous system (CNS) axons contributes to persisting neurological dysfunction after spinal cord injury (SCI). Peripheral nerve grafts (PNGs) support regeneration by thousands of injured intraspinal axons and help them bypass some of the extracellular barriers that form after SCI. However this number represents but a small portion of the total number of axons that are injured. Here we tested if rhythmic sensory stimulation during cycling exercise would boost the intrinsic regenerative state of neurons to enhance axon regeneration into PNGs after a lower thoracic (T12) spinal transection of adult rats. Using True Blue retrograde tracing, we show that 4 weeks of cycling improves regeneration into a PNG from lumbar interneurons but not by primary sensory neurons. The majority of neurons that regenerate their axon are within 5 mm of the lesion and their number increased 70% with exercise. Importantly propriospinal neurons in more distant regions (5-20 mm from the lesion) that routinely exhibit very limited regeneration responded to exercise by increasing the number of regenerating neurons by 900%. There was no exercise-associated increase in regeneration from sensory neurons. Analyses using fluorescent in situ hybridization showed that this increase in regenerative response is associated with changes in levels of mRNAs encoding the regeneration associated genes (RAGs) GAP43, β-actin and Neuritin. While propriospinal neurons showed increased mRNA levels in response to SCI alone and then to grafting and exercise, sensory neurons did not respond to SCI, but there was a response to the presence of a PNG. Thus, exercise is a non-invasive approach to modulate gene expression in injured neurons leading to an increase in regeneration. This sets the stage for future studies to test whether exercise will promote axon outgrowth beyond the PNG and reconnection with spinal cord neurons, thereby demonstrating a potential clinical application of this combined therapeutic intervention.
Collapse
Affiliation(s)
- Rahul Sachdeva
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Catherine C Theisen
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Vinu Ninan
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Jeffery L Twiss
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - John D Houlé
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|
33
|
Sharma TP, Liu Y, Wordinger RJ, Pang IH, Clark AF. Neuritin 1 promotes retinal ganglion cell survival and axonal regeneration following optic nerve crush. Cell Death Dis 2015; 6:e1661. [PMID: 25719245 PMCID: PMC4669798 DOI: 10.1038/cddis.2015.22] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 12/11/2014] [Accepted: 01/06/2015] [Indexed: 12/16/2022]
Abstract
Neuritin 1 (Nrn1) is an extracellular glycophosphatidylinositol-linked protein that stimulates axonal plasticity, dendritic arborization and synapse maturation in the central nervous system (CNS). The purpose of this study was to evaluate the neuroprotective and axogenic properties of Nrn1 on axotomized retinal ganglion cells (RGCs) in vitro and on the in vivo optic nerve crush (ONC) mouse model. Axotomized cultured RGCs treated with recombinant hNRN1 significantly increased survival of RGCs by 21% (n=6–7, P<0.01) and neurite outgrowth in RGCs by 141% compared to controls (n=15, P<0.05). RGC transduction with AAV2-CAG–hNRN1 prior to ONC promoted RGC survival (450%, n=3–7, P<0.05) and significantly preserved RGC function by 70% until 28 days post crush (dpc) (n=6, P<0.05) compared with the control AAV2-CAG–green fluorescent protein transduction group. Significantly elevated levels of RGC marker, RNA binding protein with multiple splicing (Rbpms; 73%, n=5–8, P<0.001) and growth cone marker, growth-associated protein 43 (Gap43; 36%, n=3, P<0.01) were observed 28 dpc in the retinas of the treatment group compared with the control group. Significant increase in Gap43 (100%, n=5–6, P<0.05) expression was observed within the optic nerves of the AAV2–hNRN1 group compared to controls. In conclusion, Nrn1 exhibited neuroprotective, regenerative effects and preserved RGC function on axotomized RGCs in vitro and after axonal injury in vivo. Nrn1 is a potential therapeutic target for CNS neurodegenerative diseases.
Collapse
Affiliation(s)
- T P Sharma
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Y Liu
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - R J Wordinger
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - I-H Pang
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA
| | - A F Clark
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
34
|
Neuritin can normalize neural deficits of Alzheimer's disease. Cell Death Dis 2014; 5:e1523. [PMID: 25393479 PMCID: PMC4260736 DOI: 10.1038/cddis.2014.478] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 10/05/2014] [Accepted: 10/09/2014] [Indexed: 12/15/2022]
Abstract
Reductions in hippocampal neurite complexity and synaptic plasticity are believed to contribute to the progressive impairment in episodic memory and the mild cognitive decline that occur particularly in the early stages of Alzheimer's disease (AD). Despite the functional and therapeutic importance for patients with AD, intervention to rescue or normalize dendritic elaboration and synaptic plasticity is scarcely provided. Here we show that overexpression of neuritin, an activity-dependent protein, promoted neurite outgrowth and maturation of synapses in parallel with enhanced basal synaptic transmission in cultured hippocampal neurons. Importantly, exogenous application of recombinant neuritin fully restored dendritic complexity as well as spine density in hippocampal neurons prepared from Tg2576 mice, whereas it did not affect neurite branching of neurons from their wild-type littermates. We also showed that soluble recombinant neuritin, when chronically infused into the brains of Tg2576 mice, normalized synaptic plasticity in acute hippocampal slices, leading to intact long-term potentiation. By revealing the protective actions of soluble neuritin against AD-related neural defects, we provide a potential therapeutic approach for patients with AD.
Collapse
|
35
|
Choi Y, Lee K, Ryu J, Kim HG, Jeong AY, Woo RS, Lee JH, Hyun JW, Hahn S, Kim JH, Kim HS. Neuritin attenuates cognitive function impairments in tg2576 mouse model of Alzheimer's disease. PLoS One 2014; 9:e104121. [PMID: 25101829 PMCID: PMC4125179 DOI: 10.1371/journal.pone.0104121] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 07/11/2014] [Indexed: 12/21/2022] Open
Abstract
Neuritin, also known as CPG15, is a neurotrophic factor that was initially discovered in a screen to identify genes involved in activity-dependent synaptic plasticity. Neuritin plays multiple roles in the process of neural development and synaptic plasticity, although its binding receptor(s) and downstream signaling effectors remain unclear. In this study, we found that the cortical and hippocampal expression of neuritin is reduced in the brains of Alzheimer's disease (AD) patients and demonstrated that viral-mediated expression of neuritin in the dentate gyrus of 13-month-old Tg2576 mice, an AD animal model, attenuated a deficit in learning and memory as assessed by a Morris water maze test. We also found that neuritin restored the reduction in dendritic spine density and the maturity of individual spines in primary hippocampal neuron cultures prepared from Tg2576 mice. It was also shown that viral-mediated expression of neuritin in the dentate gyrus of 7-week-old Sprague-Dawley rats increased neurogenesis in the hippocampus. Taken together, our results demonstrate that neuritin restores the reduction in dendritic spine density and the maturity of individual spines in primary hippocampal neurons from Tg2576 neurons, and also attenuates cognitive function deficits in Tg2576 mouse model of AD, suggesting that neuritin possesses a therapeutic potential for AD.
Collapse
Affiliation(s)
- Yoori Choi
- Department of Pharmacology and Biomedical Sciences, Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kihwan Lee
- Department of Pharmacology and Biomedical Sciences, Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
- National Research Laboratory for Pain, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Junghwa Ryu
- Department of Pharmacology and Biomedical Sciences, Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyoun Geun Kim
- Department of Life Science, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - A Young Jeong
- Department of Life Science, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Jun-Ho Lee
- Department of Emergency Medical Technology, Daejeon University, Daejeon, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Seokyung Hahn
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Science, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- * E-mail: (J-HK); (H-SK)
| | - Hye-Sun Kim
- Department of Pharmacology and Biomedical Sciences, Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Seoul National University Bundang Hospital, Seoul National University College of Medicine, Sungnam, Republic of Korea
- * E-mail: (J-HK); (H-SK)
| |
Collapse
|
36
|
Gao R, Wang L, Sun J, Nie K, Jian H, Gao L, Liao X, Zhang H, Huang J, Gan S. MiR-204 promotes apoptosis in oxidative stress-induced rat Schwann cells by suppressing neuritin
expression. FEBS Lett 2014; 588:3225-32. [DOI: 10.1016/j.febslet.2014.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/29/2014] [Accepted: 07/06/2014] [Indexed: 02/04/2023]
|
37
|
Ghiretti AE, Paradis S. Molecular mechanisms of activity-dependent changes in dendritic morphology: role of RGK proteins. Trends Neurosci 2014; 37:399-407. [PMID: 24910262 PMCID: PMC4113564 DOI: 10.1016/j.tins.2014.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/09/2014] [Accepted: 05/13/2014] [Indexed: 01/10/2023]
Abstract
The nervous system has the amazing capacity to transform sensory experience from the environment into changes in neuronal activity that, in turn, cause long-lasting alterations in neuronal morphology. Recent findings indicate that, surprisingly, sensory experience concurrently activates molecular signaling pathways that both promote and inhibit dendritic complexity. Historically, a number of positive regulators of activity-dependent dendritic complexity have been described, whereas the list of identified negative regulators of this process is much shorter. In recent years, there has been an emerging appreciation of the importance of the Rad/Rem/Rem2/Gem/Kir (RGK) GTPases as mediators of activity-dependent structural plasticity. In the following review, we discuss the traditional view of RGK proteins, as well as our evolving understanding of the role of these proteins in instructing structural plasticity.
Collapse
Affiliation(s)
- Amy E Ghiretti
- Department of Biology, National Center for Behavioral Genomics, and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02454, USA
| | - Suzanne Paradis
- Department of Biology, National Center for Behavioral Genomics, and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02454, USA.
| |
Collapse
|
38
|
Aberrant development and plasticity of excitatory visual cortical networks in the absence of cpg15. J Neurosci 2014; 34:3517-22. [PMID: 24599452 DOI: 10.1523/jneurosci.2955-13.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
During development, experience plays a crucial role in sculpting neuronal connections. Patterned neural activity guides formation of functional neural circuits through the selective stabilization of some synapses and the pruning of others. Activity-regulated factors are fundamental to this process, but their roles in synapse stabilization and maturation is still poorly understood. CPG15, encoded by the activity-regulated gene candidate plasticity gene 15, is a small, glycosylphosphatidylinositol (GPI)-linked, extracellular protein that promotes synapse stabilization. Here we show that global knock-out of cpg15 results in abnormal postnatal development of the excitatory network in visual cortex and an associated disruption in development of visual receptive field properties. In addition, whereas repeated stimulation induced potentiation and depression in wild-type mice, the depression was slower in cpg15 knock-out mice, suggesting impairment in short-term depression-like mechanisms. These findings establish the requirement for cpg15 in activity-dependent development of the visual system and demonstrate the importance of timely excitatory network development for normal visual function.
Collapse
|
39
|
Sharma TP, McDowell CM, Liu Y, Wagner AH, Thole D, Faga BP, Wordinger RJ, Braun TA, Clark AF. Optic nerve crush induces spatial and temporal gene expression patterns in retina and optic nerve of BALB/cJ mice. Mol Neurodegener 2014; 9:14. [PMID: 24767545 PMCID: PMC4113182 DOI: 10.1186/1750-1326-9-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 04/18/2014] [Indexed: 12/18/2022] Open
Abstract
Background Central nervous system (CNS) trauma and neurodegenerative disorders trigger a cascade of cellular and molecular events resulting in neuronal apoptosis and regenerative failure. The pathogenic mechanisms and gene expression changes associated with these detrimental events can be effectively studied using a rodent optic nerve crush (ONC) model. The purpose of this study was to use a mouse ONC model to: (a) evaluate changes in retina and optic nerve (ON) gene expression, (b) identify neurodegenerative pathogenic pathways and (c) discover potential new therapeutic targets. Results Only 54% of total neurons survived in the ganglion cell layer (GCL) 28 days post crush. Using Bayesian Estimation of Temporal Regulation (BETR) gene expression analysis, we identified significantly altered expression of 1,723 and 2,110 genes in the retina and ON, respectively. Meta-analysis of altered gene expression (≥1.5, ≤-1.5, p < 0.05) using Partek and DAVID demonstrated 28 up and 20 down-regulated retinal gene clusters and 57 up and 41 down-regulated optic nerve clusters. Regulated gene clusters included regenerative change, synaptic plasticity, axonogenesis, neuron projection, and neuron differentiation. Expression of selected genes (Vsnl1, Syt1, Synpr and Nrn1) from retinal and ON neuronal clusters were quantitatively and qualitatively examined for their relation to axonal neurodegeneration by immunohistochemistry and qRT-PCR. Conclusion A number of detrimental gene expression changes occur that contribute to trauma-induced neurodegeneration after injury to ON axons. Nrn1 (synaptic plasticity gene), Synpr and Syt1 (synaptic vesicle fusion genes), and Vsnl1 (neuron differentiation associated gene) were a few of the potentially unique genes identified that were down-regulated spatially and temporally in our rodent ONC model. Bioinformatic meta-analysis identified significant tissue-specific and time-dependent gene clusters associated with regenerative changes, synaptic plasticity, axonogenesis, neuron projection, and neuron differentiation. These ONC induced neuronal loss and regenerative failure associated clusters can be extrapolated to changes occurring in other forms of CNS trauma or in clinical neurodegenerative pathological settings. In conclusion, this study identified potential therapeutic targets to address two key mechanisms of CNS trauma and neurodegeneration: neuronal loss and regenerative failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Abbot F Clark
- North Texas Eye Research Institute, Ft, Worth, TX USA.
| |
Collapse
|
40
|
He Y, Yang G, Wang Y, Ren Y, He** X, Zhang X, Fei Z. Expression of candidate plasticity-related gene 15 is increased following traumatic brain injury. Neurol Res 2013; 35:174-80. [PMID: 23336599 DOI: 10.1179/1743132812y.0000000134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Yalong He
- Department of NeurosurgeryTeaching and Research Administration, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Guitao Yang
- Department of MedicalTeaching and Research Administration, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yangang Wang
- Department of NeurosurgeryTeaching and Research Administration, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yi Ren
- Neurosciences Graduate ProgramUniversity of Rochester Medical Center, Rochester, NY, USA
| | - Xiaosheng He**
- Department of NeurosurgeryTeaching and Research Administration, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiang Zhang
- Department of NeurosurgeryTeaching and Research Administration, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhou Fei
- Department of NeurosurgeryTeaching and Research Administration, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
41
|
Regulation of glutamate receptor internalization by the spine cytoskeleton is mediated by its PKA-dependent association with CPG2. Proc Natl Acad Sci U S A 2013; 110:E4548-56. [PMID: 24191017 DOI: 10.1073/pnas.1318860110] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
A key neuronal mechanism for adjusting excitatory synaptic strength is clathrin-mediated endocytosis of postsynaptic glutamate receptors (GluRs). The actin cytoskeleton is critical for clathrin-mediated endocytosis, yet we lack a mechanistic understanding of its interaction with the endocytic process and how it may be regulated. Here we show that F-actin in dendritic spines physically binds the synaptic nuclear envelope 1 gene product candidate plasticity gene 2 (CPG2) in a PKA-dependent manner, and that this association is required for synaptic GluR internalization. Mutating two PKA sites on CPG2 disrupts its cytoskeletal association, attenuating GluR endocytosis and affecting the efficacy of synaptic transmission in vivo. These results identify CPG2 as an F-actin binding partner that functionally mediates interaction of the spine cytoskeleton with postsynaptic endocytosis. Further, the regulation of CPG2/F-actin association by PKA provides a gateway for cellular control of synaptic receptor internalization through second messenger signaling pathways. Recent identification of human synaptic nuclear envelope 1 as a risk locus for bipolar disorder suggests that CPG2 could play a role in synaptic dysfunction underlying neuropsychiatric disease.
Collapse
|
42
|
Axonal localization of neuritin/CPG15 mRNA in neuronal populations through distinct 5' and 3' UTR elements. J Neurosci 2013; 33:13735-42. [PMID: 23966695 DOI: 10.1523/jneurosci.0962-13.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Many neuronal mRNAs are actively transported into distal axons. The 3' untranslated regions (UTRs) of axonal mRNAs often contain cues for their localization. The 3' UTR of neuritin mRNA was shown to be sufficient for localization into axons of hippocampal neurons. Here, we show that neuritin mRNA localizes into axons of rat sensory neurons, but this is predominantly driven by the 5' rather than 3' UTR. Neuritin mRNA shifts from cell body to axon predominantly after nerve crush injury, suggesting that it encodes a growth-associated protein. Consistent with this, overexpression of neuritin increases axon growth but only when its mRNA localizes into the axons.
Collapse
|
43
|
Abstract
Nuclear envelope
spectrin-repeat
proteins (Nesprins), are a novel family of
nuclear and cytoskeletal proteins with rapidly expanding roles as intracellular scaffolds
and linkers. Originally described as proteins that localise to the nuclear envelope (NE)
and establish nuclear-cytoskeletal connections, nesprins have now been found to comprise a
diverse spectrum of tissue specific isoforms that localise to multiple sub-cellular
compartments. Here, we describe how nesprins are necessary in maintaining cellular
architecture by acting as essential scaffolds and linkers at both the NE and other
sub-cellular domains. More importantly, we speculate how nesprin mutations may disrupt
tissue specific nesprin scaffolds and explain the tissue specific nature of many
nesprin-associated diseases, including laminopathies.
Collapse
|
44
|
Zheng Y, Zha Y, Spaapen RM, Mathew R, Barr K, Bendelac A, Gajewski TF. Egr2-dependent gene expression profiling and ChIP-Seq reveal novel biologic targets in T cell anergy. Mol Immunol 2013; 55:283-91. [PMID: 23548837 DOI: 10.1016/j.molimm.2013.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 03/03/2013] [Accepted: 03/04/2013] [Indexed: 02/06/2023]
Abstract
T cell anergy is one of the mechanisms contributing to peripheral tolerance, particularly in the context of progressively growing tumors and in tolerogenic treatments promoting allograft acceptance. We recently reported that early growth response gene 2 (Egr2) is a critical transcription factor for the induction of anergy in vitro and in vivo, which was identified based on its ability to regulate the expression of inhibitory signaling molecules diacylglycerol kinase (DGK)-α and -ζ. We reasoned that other transcriptional targets of Egr2 might encode additional factors important for T cell anergy and immune regulation. Thus, we conducted two sets of genome-wide screens: gene expression profiling of wild type versus Egr2-deleted T cells treated under anergizing conditions, and a ChIP-Seq analysis to identify genes that bind Egr2 in anergic cells. Merging of these data sets revealed 49 targets that are directly regulated by Egr2. Among these are inhibitory signaling molecules previously reported to contribute to T cell anergy, but unexpectedly, also cell surface molecules and secreted factors, including lymphocyte-activation gene 3 (Lag3), Class-I-MHC-restricted T cell associated molecule (Crtam), Semaphorin 7A (Sema7A), and chemokine CCL1. These observations suggest that anergic T cells might not simply be functionally inert, and may have additional functional properties oriented towards other cellular components of the immune system.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Zito A, Cartelli D, Cappelletti G, Cariboni A, Andrews W, Parnavelas J, Poletti A, Galbiati M. Neuritin 1 promotes neuronal migration. Brain Struct Funct 2012; 219:105-18. [DOI: 10.1007/s00429-012-0487-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 11/16/2012] [Indexed: 11/30/2022]
|
46
|
Rajgor D, Mellad JA, Autore F, Zhang Q, Shanahan CM. Multiple novel nesprin-1 and nesprin-2 variants act as versatile tissue-specific intracellular scaffolds. PLoS One 2012; 7:e40098. [PMID: 22768332 PMCID: PMC3388047 DOI: 10.1371/journal.pone.0040098] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 05/31/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Nesprins (Nuclear envelope spectrin-repeat proteins) are a novel family of giant spectrin-repeat containing proteins. The nesprin-1 and nesprin-2 genes consist of 146 and 116 exons which encode proteins of ∼1mDa and ∼800 kDa is size respectively when all the exons are utilised in translation. However emerging data suggests that the nesprins have multiple alternative start and termination sites throughout their genes allowing the generation of smaller isoforms. RESULTS In this study we set out to identify novel alternatively transcribed nesprin variants by screening the EST database and by using RACE analysis to identify cDNA ends. These two methods provided potential hits for alternative start and termination sites that were validated by PCR and DNA sequencing. We show that these alternative sites are not only expressed in a tissue specific manner but by combining different sites together it is possible to create a wide array of nesprin variants. By cloning and expressing small novel nesprin variants into human fibroblasts and U2OS cells we show localization to actin stress-fibres, focal adhesions, microtubules, the nucleolus, nuclear matrix and the nuclear envelope (NE). Furthermore we show that the sub-cellular localization of individual nesprin variants can vary depending on the cell type, suggesting any single nesprin variant may have different functions in different cell types. CONCLUSIONS These studies suggest nesprins act as highly versatile tissue specific intracellular protein scaffolds and identify potential novel functions for nesprins beyond cytoplasmic-nuclear coupling. These alternate functions may also account for the diverse range of disease phenotypes observed when these genes are mutated.
Collapse
Affiliation(s)
- Dipen Rajgor
- Cardiovascular Division, James Black Centre, King’s College London, London, United Kingdom
| | - Jason A. Mellad
- Cardiovascular Division, James Black Centre, King’s College London, London, United Kingdom
| | - Flavia Autore
- The Randall Division of Cell and Molecular Biophysics, New Hunt’s House, King’s College London, London, United Kingdom
| | - Qiuping Zhang
- Cardiovascular Division, James Black Centre, King’s College London, London, United Kingdom
| | - Catherine M. Shanahan
- Cardiovascular Division, James Black Centre, King’s College London, London, United Kingdom
| |
Collapse
|
47
|
Cytoskeletal interactions at the nuclear envelope mediated by nesprins. Int J Cell Biol 2012; 2012:736524. [PMID: 22518138 PMCID: PMC3296292 DOI: 10.1155/2012/736524] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 10/13/2011] [Accepted: 10/18/2011] [Indexed: 11/29/2022] Open
Abstract
Nesprin-1 is a giant tail-anchored nuclear envelope protein composed of an N-terminal F-actin binding domain, a long linker region formed by multiple spectrin repeats and a C-terminal transmembrane domain. Based on this structure, it connects the nucleus to the actin cytoskeleton. Earlier reports had shown that Nesprin-1 binds to nuclear envelope proteins emerin and lamin through C-terminal spectrin repeats. These repeats can also self-associate. We focus on the N-terminal Nesprin-1 sequences and show that they interact with Nesprin-3, a further member of the Nesprin family, which connects the nucleus to the intermediate filament network. We show that upon ectopic expression of Nesprin-3 in COS7 cells, which are nearly devoid of Nesprin-3 in vitro, vimentin filaments are recruited to the nucleus and provide evidence for an F-actin interaction of Nesprin-3 in vitro. We propose that Nesprins through interactions amongst themselves and amongst the various Nesprins form a network around the nucleus and connect the nucleus to several cytoskeletal networks of the cell.
Collapse
|
48
|
Fujino T, Leslie JH, Eavri R, Chen JL, Lin WC, Flanders GH, Borok E, Horvath TL, Nedivi E. CPG15 regulates synapse stability in the developing and adult brain. Genes Dev 2012; 25:2674-85. [PMID: 22190461 DOI: 10.1101/gad.176172.111] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Use-dependent selection of optimal connections is a key feature of neural circuit development and, in the mature brain, underlies functional adaptation, such as is required for learning and memory. Activity patterns guide circuit refinement through selective stabilization or elimination of specific neuronal branches and synapses. The molecular signals that mediate activity-dependent synapse and arbor stabilization and maintenance remain elusive. We report that knockout of the activity-regulated gene cpg15 in mice delays developmental maturation of axonal and dendritic arbors visualized by anterograde tracing and diolistic labeling, respectively. Electrophysiology shows that synaptic maturation is also delayed, and electron microscopy confirms that many dendritic spines initially lack functional synaptic contacts. While circuits eventually develop, in vivo imaging reveals that spine maintenance is compromised in the adult, leading to a gradual attrition in spine numbers. Loss of cpg15 also results in poor learning. cpg15 knockout mice require more trails to learn, but once they learn, memories are retained. Our findings suggest that CPG15 acts to stabilize active synapses on dendritic spines, resulting in selective spine and arbor stabilization and synaptic maturation, and that synapse stabilization mediated by CPG15 is critical for efficient learning.
Collapse
Affiliation(s)
- Tadahiro Fujino
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yang CB, Zheng YT, Kiser PJ, Mower GD. Identification of α-Chimaerin as a Candidate Gene for Critical Period Neuronal Plasticity in Cat and Mouse Visual Cortex. BMC Neurosci 2011; 12:70. [PMID: 21767388 PMCID: PMC3155490 DOI: 10.1186/1471-2202-12-70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 07/18/2011] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
In cat visual cortex, critical period neuronal plasticity is minimal until approximately 3 postnatal weeks, peaks at 5 weeks, gradually declines to low levels at 20 weeks, and disappears by 1 year of age. Dark rearing slows the entire time course of this critical period, such that at 5 weeks of age, normal cats are more plastic than dark reared cats, whereas at 20 weeks, dark reared cats are more plastic. Thus, a stringent criterion for identifying genes that are important for plasticity in visual cortex is that they show differences in expression between normal and dark reared that are of opposite direction in young versus older animals.
Results
The present study reports the identification by differential display PCR of a novel gene, α-chimaerin, as a candidate visual cortex critical period plasticity gene that showed bidirectional regulation of expression due to age and dark rearing. Northern blotting confirmed the bidirectional expression and 5'RACE sequencing identified the gene. There are two alternatively-spliced α-chimaerin isoforms: α1 and α2. Western blotting extended the evidence for bidirectional regulation of visual cortex α-chimaerin isoform expression to protein in cats and mice. α1- and α2-Chimaerin were elevated in dark reared compared to normal visual cortex at the peak of the normal critical period and in normal compared to dark reared visual cortex at the nadir of the normal critical period. Analysis of variance showed a significant interaction in both cats and mice for both α-chimaerin isoforms, indicating that the effect of dark rearing depended on age. This differential expression was not found in frontal cortex.
Conclusions
Chimaerins are RhoGTPase-activating proteins that are EphA4 effectors and have been implicated in a number of processes including growth cone collapse, axon guidance, dendritic spine development and the formation of corticospinal motor circuits. The present results identify α-chimaerin as a candidate molecule for a role in the postnatal critical period of visual cortical plasticity.
Collapse
|
50
|
Interaction of survival of motor neuron (SMN) and HuD proteins with mRNA cpg15 rescues motor neuron axonal deficits. Proc Natl Acad Sci U S A 2011; 108:10337-42. [PMID: 21652774 DOI: 10.1073/pnas.1104928108] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Spinal muscular atrophy (SMA), caused by the deletion of the SMN1 gene, is the leading genetic cause of infant mortality. SMN protein is present at high levels in both axons and growth cones, and loss of its function disrupts axonal extension and pathfinding. SMN is known to associate with the RNA-binding protein hnRNP-R, and together they are responsible for the transport and/or local translation of β-actin mRNA in the growth cones of motor neurons. However, the full complement of SMN-interacting proteins in neurons remains unknown. Here we used mass spectrometry to identify HuD as a novel neuronal SMN-interacting partner. HuD is a neuron-specific RNA-binding protein that interacts with mRNAs, including candidate plasticity-related gene 15 (cpg15). We show that SMN and HuD form a complex in spinal motor axons, and that both interact with cpg15 mRNA in neurons. CPG15 is highly expressed in the developing ventral spinal cord and can promote motor axon branching and neuromuscular synapse formation, suggesting a crucial role in the development of motor axons and neuromuscular junctions. Cpg15 mRNA previously has been shown to localize into axonal processes. Here we show that SMN deficiency reduces cpg15 mRNA levels in neurons, and, more importantly, cpg15 overexpression partially rescues the SMN-deficiency phenotype in zebrafish. Our results provide insight into the function of SMN protein in axons and also identify potential targets for the study of mechanisms that lead to the SMA pathology and related neuromuscular diseases.
Collapse
|