17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Neuroligin 3 Regulates Dendritic Outgrowth by Modulating Akt/mTOR Signaling

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Neuroligins (NLs) are a group of postsynaptic cell adhesion molecules that function in synaptogenesis and synaptic transmission. Genetic defects in neuroligin 3 (NL3), a member of the NL protein family, are associated with autism. Studies in rodents have revealed that mutations of NL3 gene lead to increased growth and complexity in dendrites in the central nervous system. However, the detailed mechanism is still unclear. In our study, we found that deficiency of NL3 led to morphological changes of the pyramidal neurons in layer II/III somatosensory cortex in mice, including enlarged somata, elongated dendritic length, and increased dendritic complexity. Knockdown of NL3 in cultured rat neurons upregulated Akt/mTOR signaling, resulting in both increased protein synthesis and dendritic growth. Treating neurons with either rapamycin to inhibit the mTOR or LY294002 to inhibit the PI3K/Akt activity rescued the morphological abnormalities resulting from either NL3 knockdown or knockout (KO). In addition, we found that the hyperactivated Akt/mTOR signaling associated with NL3 defects was mediated by a reduction in phosphatase and tensin (PTEN) expression, and that MAGI-2, a scaffold protein, interacted with both NL3 and PTEN and could be a linker between NL3 and Akt/mTOR signaling pathway. In conclusion, our results suggest that NL3 regulates neuronal morphology, especially dendritic outgrowth, by modulating the PTEN/Akt/mTOR signaling pathway, probably via MAGI-2. Thereby, this study provides a new link between NL3 and neuronal morphology.

          Related collections

          Most cited references60

          • Record: found
          • Abstract: found
          • Article: not found

          Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism.

          Many studies have supported a genetic etiology for autism. Here we report mutations in two X-linked genes encoding neuroligins NLGN3 and NLGN4 in siblings with autism-spectrum disorders. These mutations affect cell-adhesion molecules localized at the synapse and suggest that a defect of synaptogenesis may predispose to autism.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            eIF4 initiation factors: effectors of mRNA recruitment to ribosomes and regulators of translation.

            Eukaryotic translation initiation factor 4F (eIF4F) is a protein complex that mediates recruitment of ribosomes to mRNA. This event is the rate-limiting step for translation under most circumstances and a primary target for translational control. Functions of the constituent proteins of eIF4F include recognition of the mRNA 5' cap structure (eIF4E), delivery of an RNA helicase to the 5' region (eIF4A), bridging of the mRNA and the ribosome (eIF4G), and circularization of the mRNA via interaction with poly(A)-binding protein (eIF4G). eIF4 activity is regulated by transcription, phosphorylation, inhibitory proteins, and proteolytic cleavage. Extracellular stimuli evoke changes in phosphorylation that influence eIF4F activity, especially through the phosphoinositide 3-kinase (PI3K) and Ras signaling pathways. Viral infection and cellular stresses also affect eIF4F function. The recent determination of the structure of eIF4E at atomic resolution has provided insight about how translation is initiated and regulated. Evidence suggests that eIF4F is also implicated in malignancy and apoptosis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A mammalian protein targeted by G1-arresting rapamycin-receptor complex.

              The structurally related natural products rapamycin and FK506 bind to the same intracellular receptor, FKBP12, yet the resulting complexes interfere with distinct signalling pathways. FKBP12-rapamycin inhibits progression through the G1 phase of the cell cycle in osteosarcoma, liver and T cells as well as in yeast, and interferes with mitogenic signalling pathways that are involved in G1 progression, namely with activation of the protein p70S6k (refs 5, 11-13) and cyclin-dependent kinases. Here we isolate a mammalian FKBP-rapamycin-associated protein (FRAP) whose binding to structural variants of rapamycin complexed to FKBP12 correlates with the ability of these ligands to inhibit cell-cycle progression. Peptide sequences from purified bovine FRAP were used to isolate a human cDNA clone that is highly related to the DRR1/TOR1 and DRR2/TOR2 gene products from Saccharomyces cerevisiae. Although it has not been previously demonstrated that either of the DRR/TOR gene products can bind the FKBP-rapamycin complex directly, these yeast genes have been genetically linked to a rapamycin-sensitive pathway and are thought to encode lipid kinases.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Cell Neurosci
                Front Cell Neurosci
                Front. Cell. Neurosci.
                Frontiers in Cellular Neuroscience
                Frontiers Media S.A.
                1662-5102
                29 November 2019
                2019
                : 13
                : 518
                Affiliations
                [1] 1Department of Rehabilitation of the Children’s Hospital , Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
                [2] 2Department of Neurobiology , NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
                [3] 3Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province , Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
                [4] 4Zhejiang University-University of Edinburgh Institute , Jiaxing, China
                [5] 5Department of Orthopaedics , The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
                [6] 6JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research , Jinan University, Guangzhou, China
                [7] 7 Division of Life Science , The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
                [8] 8Division of Biomedical Engineering , The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
                [9] 9State Key Laboratory of Molecular Neuroscience , The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
                Author notes

                Edited by: Tommaso Pizzorusso, University of Florence, Italy

                Reviewed by: Claire Cheetham, University of Pittsburgh, United States; Christina Gross, Cincinnati Children’s Hospital Medical Center, United States; Eunchai Kang, University of Pennsylvania, United States

                *Correspondence: Ziyi Wang, mtray@ 123456qq.com

                This article was submitted to Cellular Neuropathology, a section of the journal Frontiers in Cellular Neuroscience

                Article
                10.3389/fncel.2019.00518
                6896717
                31849609
                ed001228-2753-4b93-af02-9fed2789ed1e
                Copyright © 2019 Xu, Du, Xu, Hu, Gu, Li, Hu, Liao, Xia, Sun, Shi, Luo, Xia, Wang and Xu.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 17 July 2019
                : 04 November 2019
                Page count
                Figures: 8, Tables: 0, Equations: 0, References: 75, Pages: 17, Words: 0
                Categories
                Neuroscience
                Original Research

                Neurosciences
                neuroligin 3,akt/mtor,dendritic outgrowth,pten,magi-2
                Neurosciences
                neuroligin 3, akt/mtor, dendritic outgrowth, pten, magi-2

                Comments

                Comment on this article