1
|
Dong A, Yoshizumi M, Kokubo H. Odz4 upregulates SAN-specific genes to promote differentiation into cardiac pacemaker-like cells. FEBS Lett 2024. [PMID: 39462648 DOI: 10.1002/1873-3468.15036] [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: 06/11/2024] [Revised: 08/03/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Cardiac arrhythmias stemming from abnormal sinoatrial node (SAN) function can lead to sudden death. Developing a biological pacemaker device for treating sick sinus syndrome (SSS) could offer a potential cure. Understanding SAN differentiation is crucial, yet its regulatory mechanism remains unclear. We reanalyzed published RNA-seq data and identified Odz4 as a SAN-specific candidate. In situ hybridization revealed Odz4 expression in the cardiac crescent and throughout the cardiac conduction system (CCS). To assess the role of Odz4 in CCS differentiation, we utilized a Tet-Off inducible system for its intracellular domain (ICD). Embryonic bodies (EBs) exogenously expressing Odz4-ICD exhibited an increased propensity to develop into pacemaker-like cells with enhanced automaticity and upregulated expression of SAN-specific genes. CellChat and GO analyses unveiled SAN-specific enrichment of ligand-receptor sets, especially Ptn-Ncl, and extracellular matrix components in the group exogenously expressing Odz4-ICD. Our findings underscore the significance of Odz4 in SAN development and offer fresh insights into biological pacemaker establishment.
Collapse
Affiliation(s)
- Anqi Dong
- Department of Physiology and Biophysics, Hiroshima University, Japan
| | - Masao Yoshizumi
- Department of Physiology and Biophysics, Hiroshima University, Japan
| | - Hiroki Kokubo
- Department of Physiology and Biophysics, Hiroshima University, Japan
- Department of Physical Therapy, Tohto University, Chiba, Japan
| |
Collapse
|
2
|
Xu C, Li Z, Lyu C, Hu Y, McLaughlin CN, Wong KKL, Xie Q, Luginbuhl DJ, Li H, Udeshi ND, Svinkina T, Mani DR, Han S, Li T, Li Y, Guajardo R, Ting AY, Carr SA, Li J, Luo L. Molecular and cellular mechanisms of teneurin signaling in synaptic partner matching. Cell 2024; 187:5081-5101.e19. [PMID: 38996528 DOI: 10.1016/j.cell.2024.06.022] [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: 02/11/2024] [Revised: 05/20/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024]
Abstract
In developing brains, axons exhibit remarkable precision in selecting synaptic partners among many non-partner cells. Evolutionarily conserved teneurins are transmembrane proteins that instruct synaptic partner matching. However, how intracellular signaling pathways execute teneurins' functions is unclear. Here, we use in situ proximity labeling to obtain the intracellular interactome of a teneurin (Ten-m) in the Drosophila brain. Genetic interaction studies using quantitative partner matching assays in both olfactory receptor neurons (ORNs) and projection neurons (PNs) reveal a common pathway: Ten-m binds to and negatively regulates a RhoGAP, thus activating the Rac1 small GTPases to promote synaptic partner matching. Developmental analyses with single-axon resolution identify the cellular mechanism of synaptic partner matching: Ten-m signaling promotes local F-actin levels and stabilizes ORN axon branches that contact partner PN dendrites. Combining spatial proteomics and high-resolution phenotypic analyses, this study advanced our understanding of both cellular and molecular mechanisms of synaptic partner matching.
Collapse
Affiliation(s)
- Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Zhuoran Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Cheng Lyu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Yixin Hu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Colleen N McLaughlin
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Kenneth Kin Lam Wong
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Qijing Xie
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - David J Luginbuhl
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Hongjie Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Namrata D Udeshi
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tanya Svinkina
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - D R Mani
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuo Han
- Departments of Genetics, Biology, and Chemistry, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA
| | - Tongchao Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Yang Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Guajardo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Alice Y Ting
- Departments of Genetics, Biology, and Chemistry, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA
| | - Steven A Carr
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jiefu Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Biology Graduate Program, Stanford University, Stanford, CA 94305, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Kurihara Y, Kawaguchi Y, Ohta Y, Kawasaki N, Fujita Y, Takei K. Nogo Receptor Antagonist LOTUS Promotes Neurite Outgrowth through Its Interaction with Teneurin-4. Cells 2024; 13:1369. [PMID: 39195260 DOI: 10.3390/cells13161369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Neurite outgrowth is a crucial process for organizing neuronal circuits in neuronal development and regeneration after injury. Regenerative failure in the adult mammalian central nervous system (CNS) is attributed to axonal growth inhibitors such as the Nogo protein that commonly binds to Nogo receptor-1 (NgR1). We previously reported that lateral olfactory tract usher substance (LOTUS) functions as an endogenous antagonist for NgR1 in forming neuronal circuits in the developing brain and improving axonal regeneration in the adult injured CNS. However, another molecular and cellular function of LOTUS remains unknown. In this study, we found that cultured retinal explant neurons extend their neurites on the LOTUS-coating substrate. This action was also observed in cultured retinal explant neurons derived from Ngr1-deficient mouse embryos, indicating that the promoting action of LOTUS on neurite outgrowth may be mediated by unidentified LOTUS-binding protein(s). We therefore screened the binding partner(s) of LOTUS by using a liquid chromatography-tandem mass spectrometry (LC-MS/MS). LC-MS/MS analysis and pull-down assay showed that LOTUS interacts with Teneurin-4 (Ten-4), a cell adhesion molecule. RNAi knockdown of Ten-4 inhibited neurite outgrowth on the LOTUS substrate in retinoic acid (RA)-treated Neuro2A cells. Furthermore, a soluble form of Ten-4 attenuates the promoting action on neurite outgrowth in cultured retinal explant neurons on the LOTUS substrate. These results suggest that LOTUS promotes neurite outgrowth by interacting with Ten-4. Our findings may provide a new molecular mechanism of LOTUS to contribute to neuronal circuit formation in development and to enhance axonal regeneration after CNS injury.
Collapse
Affiliation(s)
- Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Anatomy & Developmental Biology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Yuki Kawaguchi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 230-0045, Japan
| | - Yuki Ohta
- Laboratory of Biopharmaceutical and Regenerative Sciences, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
| | - Nana Kawasaki
- Laboratory of Biopharmaceutical and Regenerative Sciences, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
| | - Yuki Fujita
- Department of Anatomy & Developmental Biology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 230-0045, Japan
| |
Collapse
|
4
|
Tu T, Shi Y, Zhou B, Wang X, Zhang W, Zhou G, Mo X, Wang W, Wu J, Liu W. Type I collagen and fibromodulin enhance the tenogenic phenotype of hASCs and their potential for tendon regeneration. NPJ Regen Med 2023; 8:67. [PMID: 38092758 PMCID: PMC10719373 DOI: 10.1038/s41536-023-00341-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/09/2023] [Indexed: 12/17/2023] Open
Abstract
Our previous work demonstrated the tendon-derived extracellular matrix (ECM) extracts as vital niches to specifically direct mesenchymal stem cells towards tenogenic differentiation. This study aims to further define the effective ECM molecules capable of teno-lineage induction on human adipose-derived stem cells (hASCs) and test their function for tendon engineering. By detecting the teno-markers expression levels in hASCs exposed to various substrate coatings, collagen I (COL1) and fibromodulin (FMOD) were identified to be the key molecules as a combination and further employed to the modification of poly(L-lactide-co-ε-caprolactone) electrospun nanoyarns, which showed advantages in inducting seeded hASCs for teno-lineage specific differentiation. Under dynamic mechanical loading, modified scaffold seeded with hASCs formed neo-tendon in vitro at the histological level and formed better tendon tissue in vivo with mature histology and enhanced mechanical properties. Primary mechanistic investigation with RNA sequencing demonstrated that the inductive mechanism of these two molecules for hASCs tenogenic differentiation was directly correlated with positive regulation of peptidase activity, regulation of cell-substrate adhesion and regulation of cytoskeletal organization. These biological processes were potentially affected by LOC101929398/has-miR-197-3p/TENM4 ceRNA regulation axis. In summary, COL1 and FMOD in combination are the major bioactive molecules in tendon ECM for likely directing tenogenic phenotype of hASCs and certainly valuable for hASCs-based tendon engineering.
Collapse
Affiliation(s)
- Tian Tu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Plastic and Aesthetic Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Yuan Shi
- Department of Burn and Plastic Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215000, China
| | - Boya Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaoyu Wang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Tissue Engineering Center of China, Shanghai, 200241, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Tissue Engineering Center of China, Shanghai, 200241, China
| | - Xiumei Mo
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Jinglei Wu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China.
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- National Tissue Engineering Center of China, Shanghai, 200241, China.
| |
Collapse
|
5
|
Huang Y, Chen Z, Xu Y, Liu L, Tang H, He L, Zhang J, Zhou H, Xu Y, Zhao J, Wu L, Xu K. Proteomic changes of the bilateral M1 and spinal cord in hemiplegic cerebral palsy mouse: Effects of constraint-induced movement therapy. Behav Brain Res 2023; 452:114583. [PMID: 37454934 DOI: 10.1016/j.bbr.2023.114583] [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: 02/01/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Hemiplegic cerebral palsy (HCP) is a non-progressive movement and posture disorder that affects one side of the body. Constraint-induced movement therapy (CIMT) can improve the hand function of children with HCP. We used label-free proteomic quantification technology to evaluate proteomic changes in the bilateral M1 and spinal cord in HCP mouse induced by hypoxia/ischemia and CIMT. Nissl staining showed reduced neuron density in the HCP mice's lesioned and contralesional M1. The rotarod test and grip strength test showed motor dysfunction in mice with HCP and improved motor ability after CIMT. A total of 5147 proteins were identified. Fifty-one, five, and sixty common differentially expressed proteins (DEPs), which were co-regulated by HCP and CIMT, were found in the lesioned M1, the contralesional M1 and the spinal cord respectively. The significant proteins included alpha-centractin, metaxin complex, PKC, septin 11, choline transporter-like proteins, protein 4.1, teneurin-4, and so on, which mainly related to synapse stability, neuronal development and maintenance, axon development, and myelin formation. The KEGG pathways of HCP-induced DEPs mainly related to lipid metabolism, synaptic remodeling, SNARE interactions in vesicular transport and axon formation. The CIMT-induced DEPs were mainly related to synaptic remodeling and axon formation in the lesioned M1 and spinal cord. This study investigated the proteomic changes of the bilateral M1 and spinal cord as well as the CIMT-induced proteomic changes in HCP mice, which might provide new insights into the therapy of HCP.
Collapse
Affiliation(s)
- Yuan Huang
- School of Medicine, South China University of Technology, Guangzhou 510655, China; Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Zhaofang Chen
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Yunxian Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Jingbo Zhang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Hongyu Zhou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Yi Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Jingyi Zhao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Lilan Wu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China
| | - Kaishou Xu
- School of Medicine, South China University of Technology, Guangzhou 510655, China; Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120 Guangzhou, China.
| |
Collapse
|
6
|
Mihailova V, Stoyanova II, Tonchev AB. Glial Populations in the Human Brain Following Ischemic Injury. Biomedicines 2023; 11:2332. [PMID: 37760773 PMCID: PMC10525766 DOI: 10.3390/biomedicines11092332] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
There is a growing interest in glial cells in the central nervous system due to their important role in maintaining brain homeostasis under physiological conditions and after injury. A significant amount of evidence has been accumulated regarding their capacity to exert either pro-inflammatory or anti-inflammatory effects under different pathological conditions. In combination with their proliferative potential, they contribute not only to the limitation of brain damage and tissue remodeling but also to neuronal repair and synaptic recovery. Moreover, reactive glial cells can modulate the processes of neurogenesis, neuronal differentiation, and migration of neurons in the existing neural circuits in the adult brain. By discovering precise signals within specific niches, the regulation of sequential processes in adult neurogenesis holds the potential to unlock strategies that can stimulate the generation of functional neurons, whether in response to injury or as a means of addressing degenerative neurological conditions. Cerebral ischemic stroke, a condition falling within the realm of acute vascular disorders affecting the circulation in the brain, stands as a prominent global cause of disability and mortality. Extensive investigations into glial plasticity and their intricate interactions with other cells in the central nervous system have predominantly relied on studies conducted on experimental animals, including rodents and primates. However, valuable insights have also been gleaned from in vivo studies involving poststroke patients, utilizing highly specialized imaging techniques. Following the attempts to map brain cells, the role of various transcription factors in modulating gene expression in response to cerebral ischemia is gaining increasing popularity. Although the results obtained thus far remain incomplete and occasionally ambiguous, they serve as a solid foundation for the development of strategies aimed at influencing the recovery process after ischemic brain injury.
Collapse
Affiliation(s)
- Victoria Mihailova
- Department of Anatomy and Cell Biology, Faculty of Medicine, Medical University Varna, 9000 Varna, Bulgaria; (I.I.S.); (A.B.T.)
| | | | | |
Collapse
|
7
|
Campero-Romero AN, Real FH, Santana-Martínez RA, Molina-Villa T, Aranda C, Ríos-Castro E, Tovar-Y-Romo LB. Extracellular vesicles from neural progenitor cells promote functional recovery after stroke in mice with pharmacological inhibition of neurogenesis. Cell Death Discov 2023; 9:272. [PMID: 37507361 PMCID: PMC10382527 DOI: 10.1038/s41420-023-01561-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/28/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) of the subventricular zone proliferate in response to ischemic stroke in the adult mouse brain. Newly generated cells have been considered to influence recovery following a stroke. However, the mechanism underlying such protection is a matter of active study since it has been thought that proliferating NPCs mediate their protective effects by secreting soluble factors that promote recovery rather than neuronal replacement in the ischemic penumbra. We tested the hypothesis that this mechanism is mediated by the secretion of multimolecular complexes in extracellular vesicles (EVs). We found that the molecular influence of oxygen and glucose-deprived (OGD) NPCs-derived EVs is very limited in improving overt neurological alterations caused by stroke compared to our recently reported astrocyte-derived EVs. However, when we inhibited the ischemia-triggered proliferation of NPCs with the chronic administration of the DNA synthesis inhibitor Ara-C, the effect of NPC-derived EVs became evident, suggesting that the endogenous protection exerted by the proliferation of NPC is mainly carried out through a mechanism that involves the intercellular communication mediated by EVs. We analyzed the proteomic content of NPC-derived EVs cargo with label-free relative abundance mass spectrometry and identified several molecular mediators of neuronal recovery within these vesicles. Our findings indicate that NPC-derived EVs are protective against the ischemic cascade activated by stroke and, thus, hold significant therapeutic potential.
Collapse
Affiliation(s)
- Aura N Campero-Romero
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Fernando H Real
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Ricardo A Santana-Martínez
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Tonatiuh Molina-Villa
- Department of Cellular and Developmental Biology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Cristina Aranda
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Emmanuel Ríos-Castro
- Unidad de Genómica, Proteómica y Metabolómica, LaNSE, Cinvestav-IPN, Ciudad de México, México
| | - Luis B Tovar-Y-Romo
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
8
|
Durán A, Priestman DA, Las Heras M, Rebolledo-Jaramillo B, Olguín V, Calderón JF, Zanlungo S, Gutiérrez J, Platt FM, Klein AD. A Mouse Systems Genetics Approach Reveals Common and Uncommon Genetic Modifiers of Hepatic Lysosomal Enzyme Activities and Glycosphingolipids. Int J Mol Sci 2023; 24:4915. [PMID: 36902345 PMCID: PMC10002577 DOI: 10.3390/ijms24054915] [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: 01/07/2023] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Identification of genetic modulators of lysosomal enzyme activities and glycosphingolipids (GSLs) may facilitate the development of therapeutics for diseases in which they participate, including Lysosomal Storage Disorders (LSDs). To this end, we used a systems genetics approach: we measured 11 hepatic lysosomal enzymes and many of their natural substrates (GSLs), followed by modifier gene mapping by GWAS and transcriptomics associations in a panel of inbred strains. Unexpectedly, most GSLs showed no association between their levels and the enzyme activity that catabolizes them. Genomic mapping identified 30 shared predicted modifier genes between the enzymes and GSLs, which are clustered in three pathways and are associated with other diseases. Surprisingly, they are regulated by ten common transcription factors, and their majority by miRNA-340p. In conclusion, we have identified novel regulators of GSL metabolism, which may serve as therapeutic targets for LSDs and may suggest the involvement of GSL metabolism in other pathologies.
Collapse
Affiliation(s)
- Anyelo Durán
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | | | - Macarena Las Heras
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Boris Rebolledo-Jaramillo
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Valeria Olguín
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Juan F. Calderón
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 7610658, Chile
| | - Silvana Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330033, Chile
| | - Jaime Gutiérrez
- Cellular Signaling and Differentiation Laboratory, School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago 7510602, Chile
| | - Frances M. Platt
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Andrés D. Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| |
Collapse
|
9
|
Chi W, Wu M, Wang HL, Wu QY, Zhang YP, Hu YN, Zhu YB, Lin XF, Chen T, Luo JW, Ruan XL, Li YF. Han family with essential tremor caused by the P421L variant of the TENM4 gene in China. Neurol Sci 2023; 44:2003-2015. [PMID: 36689009 DOI: 10.1007/s10072-023-06603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023]
Abstract
BACKGROUND Essential tremor (ET) is an autosomal dominant inheritance disorder. Mutations in fusion sarcoma (FUS), mitochondrial serine peptidase 2 (HTRA2), teneurin transmembrane protein 4 (TENM4), sortilin1 (SORT1), SCN11A, and notch2N-terminal-like (NOTCH2NLC) genes are associated with familial ET. METHODS A proband with ET was tested using whole-exome sequencing and repeat-primed polymerase chain reaction. Subsequently, the family members were screened for the suspected mutation, and the results were verified using Sanger sequencing. The relationship between pedigree and phenotype was also analyzed, and structural and functional changes in the variants were predicted using bioinformatics analysis. RESULTS In a family with ET, the proband (III4) and the proband's father (II1), grandfather (I1), uncle (II2), and cousin (III5) all presented with involuntary tremors of both upper limbs. The responsible mutation was identified as TENM4 c.1262C > T (p.P421L), which showed genetic co-segregation in the family survey. AlphaFold predicted a change in the spatial position of TENM4 after the P421L mutation, which may have affected its stability. AlphaFold also predicted P421L to be a deleterious variation, which would lead to lower degrees of freedom of the TENM4 protein, thereby affecting the protein's structure and stability. According to the bioinformatics analysis, TENM4 (p.P421L) may reduce the signal reaching the nucleus by affecting the expression of TENM4 messenger RNA (mRNA), thereby impairing the normal oligodendrocyte differentiation process and leading to impaired myelination. CONCLUSION This study revealed that the TENM4 (p.P421L) pathogenic missense variation was responsible for ET in the proband.
Collapse
Affiliation(s)
- Wu Chi
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.,Emergency Department, Fujian Provincial Hospital, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Institute of Emergency MedicineFujian Emergency Medical Center, Fuzhou, 350001, China
| | - Min Wu
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Han-Lu Wang
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Qiu-Yan Wu
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Yan-Ping Zhang
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Ya-Nan Hu
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Yao-Bin Zhu
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.,Department of Traditional Chinese Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Xin-Fu Lin
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.,Department of Pediatrics, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Ting Chen
- Department of Traditional Chinese Medicine, Fujian Provincial Hospital, Fuzhou, 350001, China.
| | - Jie-Wei Luo
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China. .,Department of Traditional Chinese Medicine, Fujian Provincial Hospital, Fuzhou, 350001, China.
| | - Xing-Lin Ruan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| | - Yun-Fei Li
- Fujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China. .,Department of Neurology, Fujian Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
10
|
Osanai Y, Yamazaki R, Shinohara Y, Ohno N. Heterogeneity and regulation of oligodendrocyte morphology. Front Cell Dev Biol 2022; 10:1030486. [PMID: 36393856 PMCID: PMC9644283 DOI: 10.3389/fcell.2022.1030486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/10/2022] [Indexed: 09/24/2023] Open
Abstract
Oligodendrocytes form multiple myelin sheaths in the central nervous system (CNS), which increase nerve conduction velocity and are necessary for basic and higher brain functions such as sensory function, motor control, and learning. Structures of the myelin sheath such as myelin internodal length and myelin thickness regulate nerve conduction. Various parts of the central nervous system exhibit different myelin structures and oligodendrocyte morphologies. Recent studies supported that oligodendrocytes are a heterogenous population of cells and myelin sheaths formed by some oligodendrocytes can be biased to particular groups of axons, and myelin structures are dynamically modulated in certain classes of neurons by specific experiences. Structures of oligodendrocyte/myelin are also affected in pathological conditions such as demyelinating and neuropsychiatric disorders. This review summarizes our understanding of heterogeneity and regulation of oligodendrocyte morphology concerning central nervous system regions, neuronal classes, experiences, diseases, and how oligodendrocytes are optimized to execute central nervous system functions.
Collapse
Affiliation(s)
- Yasuyuki Osanai
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Reiji Yamazaki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yoshiaki Shinohara
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
11
|
Kwak MJ, Kim WY, Jung SH, Chung YJ, Kim JH. Differential transcriptome profile underlying risky choice in a rat gambling task. J Behav Addict 2022; 11:845-857. [PMID: 36094860 PMCID: PMC9872528 DOI: 10.1556/2006.2022.00068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/20/2022] [Accepted: 08/22/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND AND AIMS Proper measurement of expected risk is important for making rational decisions, and maladaptive decision making may underlie various psychiatric disorders. However, differentially expressed genetic profiling involved in this process is still largely unknown. A rodent version of the gambling task (rGT) has been developed to measure decision-making by adopting the same principle of Iowa Gambling Task in humans. In the present study, we examined using next-generation sequencing (NGS) technique whether there are differences in gene expression profiles in the medial prefrontal cortex (mPFC) and the nucleus accumbens (NAc) when rats make different choices toward risk in rGT. METHODS Rats were trained in a touch screen chamber to learn the relationships between 4 different light signals on the window of the screen and accompanied reward outcomes or punishments set up with different magnitudes and probabilities. Once they showed a stabilized pattern of preference upon free choice, rats were classified into risk-averse or risk-seeking groups. After performing the rGT, rats were decapitated, the mPFC and the NAc was dissected out, and NGS was performed with the total RNA extracted. RESULTS We found that 477 and 36 genes were differentially expressed (approximately 75 and 83% out of them were downregulated) in the mPFC and the NAc, respectively, in risk-seeking compared to risk-averse rats. Among those, we suggested a few top ranked genes that may contribute to promoting risky choices. DISCUSSION AND CONCLUSIONS Our findings provide insights into transcriptional components underlying risky choices in rats.
Collapse
Affiliation(s)
- Myung Ji Kwak
- Department of Medical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Wha Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Seung-Hyun Jung
- Department of Biochemistry, Cancer Evolution Research Center, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea,Department of Biomedicine & Health Sciences, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea,Precision Medicine Research Center, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea,Corresponding authors. E-mail: , ,
| | - Yeun-Jun Chung
- Department of Biomedicine & Health Sciences, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea,Precision Medicine Research Center, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea,Department of Microbiology, IRCGP, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea,Corresponding authors. E-mail: , ,
| | - Jeong-Hoon Kim
- Department of Medical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea,Department of Physiology, Yonsei University College of Medicine, Seoul 03722, South Korea,Corresponding authors. E-mail: , ,
| |
Collapse
|
12
|
Yamada M, Iwase M, Sasaki B, Suzuki N. The molecular regulation of oligodendrocyte development and CNS myelination by ECM proteins. Front Cell Dev Biol 2022; 10:952135. [PMID: 36147746 PMCID: PMC9488109 DOI: 10.3389/fcell.2022.952135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Oligodendrocytes are myelin-forming cells in the central nervous system (CNS). The development of oligodendrocytes is regulated by a large number of molecules, including extracellular matrix (ECM) proteins that are relatively less characterized. Here, we review the molecular functions of the major ECM proteins in oligodendrocyte development and pathology. Among the ECM proteins, laminins are positive regulators in oligodendrocyte survival, differentiation, and/or myelination in the CNS. Conversely, fibronectin, tenascin-C, hyaluronan, and chondroitin sulfate proteoglycans suppress the differentiation and myelination. Tenascin-R shows either positive or negative functions in these activities. In addition, the extracellular domain of the transmembrane protein teneurin-4, which possesses the sequence homology with tenascins, promotes the differentiation of oligodendrocytes. The activities of these ECM proteins are exerted through binding to the cellular receptors and co-receptors, such as integrins and growth factor receptors, which induces the signaling to form the elaborated and functional structure of myelin. Further, the ECM proteins dynamically change their structures and functions at the pathological conditions as multiple sclerosis. The ECM proteins are a critical player to serve as a component of the microenvironment for oligodendrocytes in their development and pathology.
Collapse
|
13
|
Phenotypes, mechanisms and therapeutics: insights from bipolar disorder GWAS findings. Mol Psychiatry 2022; 27:2927-2939. [PMID: 35351989 DOI: 10.1038/s41380-022-01523-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 12/25/2022]
Abstract
Genome-wide association studies (GWAS) have reported substantial genomic loci significantly associated with clinical risk of bipolar disorder (BD), and studies combining techniques of genetics, neuroscience, neuroimaging, and pharmacology are believed to help tackle clinical problems (e.g., identifying novel therapeutic targets). However, translating findings of psychiatric genetics into biological mechanisms underlying BD pathogenesis remains less successful. Biological impacts of majority of BD GWAS risk loci are obscure, and the involvement of many GWAS risk genes in this illness is yet to be investigated. It is thus necessary to review the progress of applying BD GWAS risk genes in the research and intervention of the disorder. A comprehensive literature search found that a number of such risk genes had been investigated in cellular or animal models, even before they were highlighted in BD GWAS. Intriguingly, manipulation of many BD risk genes (e.g., ANK3, CACNA1C, CACNA1B, HOMER1, KCNB1, MCHR1, NCAN, SHANK2 etc.) resulted in altered murine behaviors largely restoring BD clinical manifestations, including mania-like symptoms such as hyperactivity, anxiolytic-like behavior, as well as antidepressant-like behavior, and these abnormalities could be attenuated by mood stabilizers. In addition to recapitulating phenotypic characteristics of BD, some GWAS risk genes further provided clues for the neurobiology of this illness, such as aberrant activation and functional connectivity of brain areas in the limbic system, and modulated dendritic spine morphogenesis as well as synaptic plasticity and transmission. Therefore, BD GWAS risk genes are undoubtedly pivotal resources for modeling this illness, and might be translational therapeutic targets in the future clinical management of BD. We discuss both promising prospects and cautions in utilizing the bulk of useful resources generated by GWAS studies. Systematic integrations of findings from genetic and neuroscience studies are called for to promote our understanding and intervention of BD.
Collapse
|
14
|
Zhang X, Lin PY, Liakath-Ali K, Südhof TC. Teneurins assemble into presynaptic nanoclusters that promote synapse formation via postsynaptic non-teneurin ligands. Nat Commun 2022; 13:2297. [PMID: 35484136 PMCID: PMC9050732 DOI: 10.1038/s41467-022-29751-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
Extensive studies concluded that homophilic interactions between pre- and postsynaptic teneurins, evolutionarily conserved cell-adhesion molecules, encode the specificity of synaptic connections. However, no direct evidence is available to demonstrate that teneurins are actually required on both pre- and postsynaptic neurons for establishing synaptic connections, nor is it known whether teneurins are localized to synapses. Using super-resolution microscopy, we demonstrate that Teneurin-3 assembles into presynaptic nanoclusters of approximately 80 nm in most excitatory synapses of the hippocampus. Presynaptic deletions of Teneurin-3 and Teneurin-4 in the medial entorhinal cortex revealed that they are required for assembly of entorhinal cortex-CA1, entorhinal cortex-subiculum, and entorhinal cortex-dentate gyrus synapses. Postsynaptic deletions of teneurins in the CA1 region, however, had no effect on synaptic connections from any presynaptic input. Our data suggest that different from the current prevailing view, teneurins promote the establishment of synaptic connections exclusively as presynaptic cell-adhesion molecules, most likely via their nanomolar-affinity binding to postsynaptic latrophilins.
Collapse
Affiliation(s)
- Xuchen Zhang
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA. .,Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
| | - Pei-Yi Lin
- grid.168010.e0000000419368956Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA USA
| | - Kif Liakath-Ali
- grid.168010.e0000000419368956Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA USA
| | - Thomas C. Südhof
- grid.168010.e0000000419368956Howard Hughes Medical Institute, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA USA
| |
Collapse
|
15
|
Meijer DH, Frias CP, Beugelink JW, Deurloo YN, Janssen BJC. Teneurin4 dimer structures reveal a calcium‐stabilized compact conformation supporting homomeric trans‐interactions. EMBO J 2022; 41:e107505. [PMID: 35099835 PMCID: PMC9058538 DOI: 10.15252/embj.2020107505] [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: 12/16/2020] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 11/18/2022] Open
Abstract
Establishment of correct synaptic connections is a crucial step during neural circuitry formation. The Teneurin family of neuronal transmembrane proteins promotes cell–cell adhesion via homophilic and heterophilic interactions, and is required for synaptic partner matching in the visual and hippocampal systems in vertebrates. It remains unclear how individual Teneurins form macromolecular cis‐ and trans‐synaptic protein complexes. Here, we present a 2.7 Å cryo‐EM structure of the dimeric ectodomain of human Teneurin4. The structure reveals a compact conformation of the dimer, stabilized by interactions mediated by the C‐rich, YD‐shell, and ABD domains. A 1.5 Å crystal structure of the C‐rich domain shows three conserved calcium binding sites, and thermal unfolding assays and SAXS‐based rigid‐body modeling demonstrate that the compactness and stability of Teneurin4 dimers are calcium‐dependent. Teneurin4 dimers form a more extended conformation in conditions that lack calcium. Cellular assays reveal that the compact cis‐dimer is compatible with homomeric trans‐interactions. Together, these findings support a role for teneurins as a scaffold for macromolecular complex assembly and the establishment of cis‐ and trans‐synaptic interactions to construct functional neuronal circuits.
Collapse
Affiliation(s)
- Dimphna H Meijer
- Department of Bionanoscience Kavli Institute of Nanoscience Delft University of Technology Delft The Netherlands
- Department of Chemistry Faculty of Science Structural Biochemistry Bijvoet Center for Biomolecular Research Utrecht University Utrecht The Netherlands
| | - Cátia P Frias
- Department of Bionanoscience Kavli Institute of Nanoscience Delft University of Technology Delft The Netherlands
| | - J Wouter Beugelink
- Department of Chemistry Faculty of Science Structural Biochemistry Bijvoet Center for Biomolecular Research Utrecht University Utrecht The Netherlands
| | - Yanthi N Deurloo
- Department of Bionanoscience Kavli Institute of Nanoscience Delft University of Technology Delft The Netherlands
| | - Bert J C Janssen
- Department of Chemistry Faculty of Science Structural Biochemistry Bijvoet Center for Biomolecular Research Utrecht University Utrecht The Netherlands
| |
Collapse
|
16
|
Role and Involvement of TENM4 and miR-708 in Breast Cancer Development and Therapy. Cells 2022; 11:cells11010172. [PMID: 35011736 PMCID: PMC8750459 DOI: 10.3390/cells11010172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/31/2021] [Accepted: 01/02/2022] [Indexed: 12/20/2022] Open
Abstract
Teneurin 4 (TENM4) is a transmembrane protein that is codified by the ODZ4 gene and is involved in nervous system development, neurite outgrowth, and neuronal differentiation. In line with its involvement in the nervous system, TENM4 has also been implicated in several mental disorders such as bipolar disorder, schizophrenia, and autism. TENM4 mutations and rearrangements have recently been identified in a number of tumors. This, combined with impaired expression in tumors, suggests that it may potentially be involved in tumorigenesis. Most of the TENM4 mutations that are observed in tumors occur in breast cancer, in which TENM4 plays a role in cells’ migration and stemness. However, the functional role that TENM4 plays in breast cancer still needs to be better evaluated, and further studies are required to better understand the involvement of TENM4 in breast cancer progression. Herein, we review the currently available data for TENM4′s role in breast cancer and propose its use as both a novel target with which to ameliorate patient prognosis and as a potential biomarker. Moreover, we also report data on the tumorigenic role of miR-708 deregulation and the possible use of this miRNA as a novel therapeutic molecule, as miR-708 is spliced out from TENM4 mRNA.
Collapse
|
17
|
Cardona HJ, Somasundaram A, Crabtree DM, Gadd SL, Becher OJ. Prenatal overexpression of platelet-derived growth factor receptor A results in central nervous system hypomyelination. Brain Behav 2021; 11:e2332. [PMID: 34480532 PMCID: PMC8553322 DOI: 10.1002/brb3.2332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/01/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Platelet-derived growth factor (PDGF) signaling, through the ligand PDGF-A and its receptor PDGFRA, is important for the growth and maintenance of oligodendrocyte progenitor cells (OPCs) in the central nervous system (CNS). PDGFRA signaling is downregulated prior to OPC differentiation into mature myelinating oligodendrocytes. By contrast, PDGFRA is often genetically amplified or mutated in many types of gliomas, including diffuse midline glioma (DMG) where OPCs are considered the most likely cell-of-origin. The cellular and molecular changes that occur in OPCs in response to unregulated PDGFRA expression, however, are not known. METHODS Here, we created a conditional knock-in (KI) mouse that overexpresses wild type (WT) human PDGFRA (hPDGFRA) in prenatal Olig2-expressing progenitors, and examined in vivo cellular and molecular consequences. RESULTS The KI mice exhibited stunted growth, ataxia, and a severe loss of myelination in the brain and spinal cord. When combined with the loss of p53, a tumor suppressor gene whose activity is decreased in DMG, the KI mice failed to develop tumors but still exhibited hypomyelination. RNA-sequencing analysis revealed decreased myelination gene signatures, indicating a defect in oligodendroglial development. Mice overexpressing PDGFRA in prenatal GFAP-expressing progenitors, which give rise to a broader lineage of cells than Olig2-progenitors, also developed myelination defects. CONCLUSION Our results suggest that embryonic overexpression of hPDGFRA in Olig2- or GFAP-progenitors is deleterious to OPC development and leads to CNS hypomyelination.
Collapse
Affiliation(s)
- Herminio Joey Cardona
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Agila Somasundaram
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Donna M Crabtree
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA.,Office of Clinical Research, Duke University Medical Center, Durham, NC, USA
| | - Samantha L Gadd
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Oren J Becher
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA.,Department of Pediatrics, Northwestern University, Chicago, Illinois, USA.,Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
18
|
Yi X, Li M, He G, Du H, Li X, Cao D, Wang L, Wu X, Yang F, Chen X, He L, Ping Y, Zhou D. Genetic and functional analysis reveals TENM4 contributes to schizophrenia. iScience 2021; 24:103063. [PMID: 34568788 PMCID: PMC8449235 DOI: 10.1016/j.isci.2021.103063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/23/2021] [Accepted: 08/26/2021] [Indexed: 12/09/2022] Open
Abstract
TENM4, encoding a member of the teneurin protein family, is a risk gene shared by many types of mental diseases and is implicated in neuronal plasticity and signaling. However, the role and the mechanisms of TENM4 in schizophrenia (SCZ) remain unclear. We identified possible pathogenic mutations in the TENM4 gene through target sequencing of TENM4 in 68 SCZ families. We further demonstrated that aberrant expression of Ten-m leads to lower learning ability, sleep reduction, and increased aggressiveness in animal models. RNA sequencing showed that aberrant expression of Ten-m was related to stimulus perception and metabolic process, and Gene Ontology enrichment terms were neurogenesis and ATPase activity. This study provides strong evidence that TENM4 contributes to SCZ, and its functional mutations might be responsible for the impaired neural circuits and behaviors observed in SCZ. Possible pathogenic rare missense mutations in TENM4 gene contribute to SCZ Aberrant expression of Ten-m leads to behavioral disturbances related to SCZ symptoms Ten-m affects stimulation, metabolic process, neurogenesis, and ATPase activity
Collapse
Affiliation(s)
- Xin Yi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Minzhe Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huihui Du
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Xingwang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongmei Cao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Lu Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Xi Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Fengping Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Xu Chen
- Department of Neurology, Shanghai Eighth People's Hospital, Shanghai Sixth People's Hospital Xuhui Branch, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Daizhan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| |
Collapse
|
19
|
Shimizu T, Murakoshi H, Matsumoto H, Ichino K, Hattori A, Ueno S, Ishida A, Tajiri N, Hida H. Tension Sensor Based on Fluorescence Resonance Energy Transfer Reveals Fiber Diameter-Dependent Mechanical Factors During Myelination. Front Cell Neurosci 2021; 15:685044. [PMID: 34408628 PMCID: PMC8364977 DOI: 10.3389/fncel.2021.685044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/13/2021] [Indexed: 11/15/2022] Open
Abstract
Oligodendrocytes (OLs) form a myelin sheath around neuronal axons to increase conduction velocity of action potential. Although both large and small diameter axons are intermingled in the central nervous system (CNS), the number of myelin wrapping is related to the axon diameter, such that the ratio of the diameter of the axon to that of the entire myelinated-axon unit is optimal for each axon, which is required for exerting higher brain functions. This indicates there are unknown axon diameter-dependent factors that control myelination. We tried to investigate physical factors to clarify the mechanisms underlying axon diameter-dependent myelination. To visualize OL-generating forces during myelination, a tension sensor based on fluorescence resonance energy transfer (FRET) was used. Polystyrene nanofibers with varying diameters similar to neuronal axons were prepared to investigate biophysical factors regulating the OL-axon interactions. We found that higher tension was generated at OL processes contacting larger diameter fibers compared with smaller diameter fibers. Additionally, OLs formed longer focal adhesions (FAs) on larger diameter axons and shorter FAs on smaller diameter axons. These results suggest that OLs respond to the fiber diameter and activate mechanotransduction initiated at FAs, which controls their cytoskeletal organization and myelin formation. This study leads to the novel and interesting idea that physical factors are involved in myelin formation in response to axon diameter.
Collapse
Affiliation(s)
- Takeshi Shimizu
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hideji Murakoshi
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Hidetoshi Matsumoto
- Department of Materials Science and Engineering, Tokyo Institute of Technology, Meguro, Japan
| | - Kota Ichino
- Department of Materials Science and Engineering, Tokyo Institute of Technology, Meguro, Japan
| | - Atsunori Hattori
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shinya Ueno
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimasa Ishida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Naoki Tajiri
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hideki Hida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
20
|
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Álvarez I, Pastor P, Agúndez JAG. Genomic Markers for Essential Tremor. Pharmaceuticals (Basel) 2021; 14:ph14060516. [PMID: 34072005 PMCID: PMC8226734 DOI: 10.3390/ph14060516] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022] Open
Abstract
There are many reports suggesting an important role of genetic factors in the etiopathogenesis of essential tremor (ET), encouraging continuing the research for possible genetic markers. Linkage studies in families with ET have identified 4 genes/loci for familial ET, although the responsible gene(s) have not been identified. Genome-wide association studies (GWAS) described several variants in LINGO1, SLC1A2, STK32B, PPARGC1A, and CTNNA3, related with ET, but none of them have been confirmed in replication studies. In addition, the case-control association studies performed for candidate variants have not convincingly linked any gene with the risk for ET. Exome studies described the association of several genes with familial ET (FUS, HTRA2, TENM4, SORT1, SCN11A, NOTCH2NLC, NOS3, KCNS2, HAPLN4, USP46, CACNA1G, SLIT3, CCDC183, MMP10, and GPR151), but they were found only in singular families and, again, not found in other families or other populations, suggesting that some can be private polymorphisms. The search for responsible genes for ET is still ongoing.
Collapse
Affiliation(s)
- Félix Javier Jiménez-Jiménez
- Section of Neurology, Hospital Universitario del Sureste, E28500 Arganda del Rey, Spain;
- Correspondence: ; Tel.: +34-636-96-83-95; Fax: +34-913-28-07-04
| | | | - Elena García-Martín
- ARADyAL Instituto de Salud Carlos III, University Institute of Molecular Pathology Biomarkers, University of Extremadura, E10071 Caceres, Spain; (E.G.-M.); (J.A.G.A.)
| | - Ignacio Álvarez
- Movement Disorders Unit, Department of Neurology, University Hospital Mútua de Terrassa, Fundació Docencia i Recerça Mútua de Terrassa, E08221 Terrassa, Spain; (I.Á.); (P.P.)
| | - Pau Pastor
- Movement Disorders Unit, Department of Neurology, University Hospital Mútua de Terrassa, Fundació Docencia i Recerça Mútua de Terrassa, E08221 Terrassa, Spain; (I.Á.); (P.P.)
| | - José A. G. Agúndez
- ARADyAL Instituto de Salud Carlos III, University Institute of Molecular Pathology Biomarkers, University of Extremadura, E10071 Caceres, Spain; (E.G.-M.); (J.A.G.A.)
| |
Collapse
|
21
|
Cortés-Montero E, Rodríguez-Muñoz M, Sánchez-Blázquez P, Garzón-Niño J. Human HINT1 Mutant Proteins that Cause Axonal Motor Neuropathy Exhibit Anomalous Interactions with Partner Proteins. Mol Neurobiol 2021; 58:1834-1845. [PMID: 33404983 DOI: 10.1007/s12035-020-02265-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/16/2020] [Indexed: 11/25/2022]
Abstract
The 14 kDa histidine triad nucleotide-binding protein 1 (HINT1) is critical to maintain the normal function of motor neurons. Thus, a series of human HINT1 mutants cause autosomal recessive axonal neuropathy with neuromyotonia. HINT1 establishes a series of regulatory interactions with signaling proteins, some of which are enriched in motor neurons, such as the type 1 sigma receptor or intracellular domain (ICD) of transmembrane teneurin 1, both of which are also implicated in motor disturbances. In a previous study, we reported the capacity of HINT1 to remove the small ubiquitin-like modifier (SUMO) from a series of substrates and the influence of HINT1 mutants on this activity. We now report how human HINT1 mutations affect the interaction of HINT1 with the regulator of its SUMOylase activity, calcium-activated calmodulin, and its substrate SUMO. Moreover, HINT1 mutants exhibited anomalous interactions with G protein coupled receptors, such as the mu-opioid, and with glutamate N-methyl-D-aspartate receptors as well. Additionally, these HINT1 mutants showed impaired associations with transcriptional regulators such as the regulator of G protein signaling Z2 protein and the cleaved N-terminal ICD of teneurin 1. Thus, the altered enzymatic activity of human HINT1 mutants and their anomalous interactions with partner proteins may disrupt signaling pathways essential to the normal function of human motor neurons.
Collapse
Affiliation(s)
- Elsa Cortés-Montero
- Neuropharmacology, Cajal Institute, Department of Translational Neuroscience, CSIC, Madrid, Spain
| | - María Rodríguez-Muñoz
- Neuropharmacology, Cajal Institute, Department of Translational Neuroscience, CSIC, Madrid, Spain
| | - Pilar Sánchez-Blázquez
- Neuropharmacology, Cajal Institute, Department of Translational Neuroscience, CSIC, Madrid, Spain
| | - Javier Garzón-Niño
- Neuropharmacology, Cajal Institute, Department of Translational Neuroscience, CSIC, Madrid, Spain.
| |
Collapse
|
22
|
Takano I, Takeshita N, Yoshida M, Seki D, Oyanagi T, Kimura S, Jiang W, Sasaki K, Sogi C, Kawatsu M, Takano-Yamamoto T. Ten-m/Odz3 regulates migration and differentiation of chondrogenic ATDC5 cells via RhoA-mediated actin reorganization. J Cell Physiol 2021; 236:2906-2919. [PMID: 32960451 DOI: 10.1002/jcp.30058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
Tenascin-like molecule major (Ten-m)/odd Oz (Odz), a type II transmembrane molecule, is well known to modulate neural development. We have reported that Ten-m/Odz3 is expressed in cartilaginous tissues and cells. Actin cytoskeleton and its regulator ras homolog gene family member A (RhoA) are closely associated with chondrogenesis. The present study aimed to evaluate the function and molecular mechanism of Ten-m/Odz3 during chondrogenesis, focusing on RhoA and the actin cytoskeleton. Ten-m/Odz3 was expressed in precartilaginous condensing mesenchyme in mouse limb buds. Ten-m/Odz3 knockdown in ATDC5 induced actin cytoskeleton reorganization and change of cell shape through modulation of RhoA activity and FGF2 expression. Ten-m/Odz3 knockdown suppressed ATDC5 migration and expression of genes associated with chondrogenesis, such as Sox9 and type II collagen, via RhoA. On the other hand, Ten-m/Odz3 knockdown inhibited proliferation of ATDC5 in a RhoA-independent manner. These findings suggest that Ten-m/Odz3 plays an important role in early chondrogenesis regulating RhoA-mediated actin reorganization.
Collapse
Affiliation(s)
- Ikuko Takano
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Nobuo Takeshita
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Michiko Yoshida
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Daisuke Seki
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Toshihito Oyanagi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Seiji Kimura
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Wei Jiang
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Kiyo Sasaki
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Chisumi Sogi
- Department of Pediatrics, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Masayoshi Kawatsu
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Teruko Takano-Yamamoto
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
- Department of Biomaterials and Bioengineering, Faculty of Dental Medicine, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
23
|
Teneurins: Role in Cancer and Potential Role as Diagnostic Biomarkers and Targets for Therapy. Int J Mol Sci 2021; 22:ijms22052321. [PMID: 33652578 PMCID: PMC7956758 DOI: 10.3390/ijms22052321] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Teneurins have been identified in vertebrates as four different genes (TENM1-4), coding for membrane proteins that are mainly involved in embryonic and neuronal development. Genetic studies have correlated them with various diseases, including developmental problems, neurological disorders and congenital general anosmia. There is some evidence to suggest their possible involvement in cancer initiation and progression, and drug resistance. Indeed, mutations, chromosomal alterations and the deregulation of teneurins expression have been associated with several tumor types and patient survival. However, the role of teneurins in cancer-related regulatory networks is not fully understood, as both a tumor-suppressor role and pro-tumoral functions have been proposed, depending on tumor histotype. Here, we summarize and discuss the literature data on teneurins expression and their potential role in different tumor types, while highlighting the possibility of using teneurins as novel molecular diagnostic and prognostic biomarkers and as targets for cancer treatments, such as immunotherapy, in some tumors.
Collapse
|
24
|
Ruiu R, Barutello G, Arigoni M, Riccardo F, Conti L, Peppino G, Annaratone L, Marchiò C, Mengozzi G, Calogero RA, Cavallo F, Quaglino E. Identification of TENM4 as a Novel Cancer Stem Cell-Associated Molecule and Potential Target in Triple Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13040894. [PMID: 33672732 PMCID: PMC7924390 DOI: 10.3390/cancers13040894] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Patients with triple negative breast cancer (TNBC) experience shorter overall survival compared to non-TNBC patients because of the high incidence of recurrences and metastases. This is due to the capacity of aggressive cancer cell subpopulations named cancer stem cells (CSC) to resist current therapies. To design more effective therapeutic strategies for TNBC patients, in this study we sought to identify functional targets expressed on CSC. Our analyses led us to propose teneurin 4 (TENM4) as a promising candidate for drug- and immune-based therapies due to its role in CSC self-renewal and migratory capacity and the inverse correlation between its expression and survival of TNBC patients. In addition, TENM4 detection in the plasma of tumor-bearing patients endorses its potentiality as a disease detection marker. Abstract Triple-negative breast cancer (TNBC) is insensitive to endocrine and Her2-directed therapies, making the development of TNBC-targeted therapies an unmet medical need. Since patients with TNBC frequently show a quicker relapse and metastatic progression compared to other breast cancer subtypes, we hypothesized that cancer stem cells (CSC) could have a role in TNBC. To identify putative TNBC CSC-associated targets, we compared the gene expression profiles of CSC-enriched tumorspheres and their parental cells grown as monolayer. Among the up-regulated genes coding for cell membrane-associated proteins, we selected Teneurin 4 (TENM4), involved in cell differentiation and deregulated in tumors of different histotypes, as the object for this study. Meta-analysis of breast cancer datasets shows that TENM4 mRNA is up-regulated in invasive carcinoma specimens compared to normal breast and that high expression of TENM4 correlates with a shorter relapse-free survival in TNBC patients. TENM4 silencing in mammary cancer cells significantly impaired tumorsphere-forming ability, migratory capacity and Focal Adhesion Kinase (FAK) phosphorylation. Moreover, we found higher levels of TENM4 in plasma from tumor-bearing mice and TNBC patients compared to the healthy controls. Overall, our results indicate that TENM4 may act as a novel biomarker and target for the treatment of TNBC.
Collapse
Affiliation(s)
- Roberto Ruiu
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
| | - Giuseppina Barutello
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
| | - Maddalena Arigoni
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
| | - Federica Riccardo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
| | - Laura Conti
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
| | - Giulia Peppino
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
| | - Laura Annaratone
- Unit of Pathology, Candiolo Cancer Institute, FPO IRCCS, 10060 Candiolo, Italy; (L.A.); (C.M.)
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy;
| | - Caterina Marchiò
- Unit of Pathology, Candiolo Cancer Institute, FPO IRCCS, 10060 Candiolo, Italy; (L.A.); (C.M.)
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy;
| | - Giulio Mengozzi
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy;
- Clinical Biochemistry Laboratory, Department of Laboratory Medicine, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Raffaele Adolfo Calogero
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
| | - Federica Cavallo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
| | - Elena Quaglino
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (R.R.); (G.B.); (M.A.); (F.R.); (L.C.); (G.P.); (R.A.C.); (F.C.)
- Correspondence: ; Tel.: +39-0116706457
| |
Collapse
|
25
|
Kim HY, Um JW, Ko J. Proper synaptic adhesion signaling in the control of neural circuit architecture and brain function. Prog Neurobiol 2021; 200:101983. [PMID: 33422662 DOI: 10.1016/j.pneurobio.2020.101983] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/23/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Trans-synaptic cell-adhesion molecules are critical for governing various stages of synapse development and specifying neural circuit properties via the formation of multifarious signaling pathways. Recent studies have pinpointed the putative roles of trans-synaptic cell-adhesion molecules in mediating various cognitive functions. Here, we review the literature on the roles of a diverse group of central synaptic organizers, including neurexins (Nrxns), leukocyte common antigen-related receptor protein tyrosine phosphatases (LAR-RPTPs), and their associated binding proteins, in regulating properties of specific type of synapses and neural circuits. In addition, we highlight the findings that aberrant synaptic adhesion signaling leads to alterations in the structures, transmission, and plasticity of specific synapses across diverse brain areas. These results seem to suggest that proper trans-synaptic signaling pathways by Nrxns, LAR-RPTPs, and their interacting network is likely to constitute central molecular complexes that form the basis for cognitive functions, and that these complexes are heterogeneously and complexly disrupted in many neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea; Core Protein Resources Center, DGIST, Daegu, 42988, South Korea.
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea.
| |
Collapse
|
26
|
Diez-Fairen M, Houle G, Ortega-Cubero S, Bandres-Ciga S, Alvarez I, Carcel M, Ibañez L, Fernandez MV, Budde JP, Trotta JR, Tonda R, Chong JX, Bamshad MJ, Nickerson DA, Aguilar M, Tartari JP, Gironell A, García-Martín E, Agundez JA, Alonso-Navarro H, Jimenez-Jimenez FJ, Fernandez M, Valldeoriola F, Marti MJ, Tolosa E, Coria F, Pastor MA, Vilariño-Güell C, Rajput A, Dion PA, Cruchaga C, Rouleau GA, Pastor P. Exome-wide rare variant analysis in familial essential tremor. Parkinsonism Relat Disord 2020; 82:109-116. [PMID: 33279834 DOI: 10.1016/j.parkreldis.2020.11.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/08/2020] [Accepted: 11/21/2020] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Essential tremor (ET) is one of the most common movement disorders. Despite its high prevalence and heritability, its genetic etiology remains elusive with only a few susceptibility genes identified and poorly replicated. Our aim was to find novel candidate genes involved in ET predisposition through whole exome sequencing. METHODS We studied eight multigenerational families (N = 40 individuals) with an autosomal-dominant inheritance using a comprehensive strategy combining whole exome sequencing followed by case-control association testing of prioritized variants in a separate cohort comprising 521 ET cases and 596 controls. We further performed gene-based burden analyses in an additional dataset comprising 789 ET patients and 770 healthy individuals to investigate whether there was an enrichment of rare deleterious variants within our candidate genes. RESULTS Fifteen variants co-segregated with disease status in at least one of the families, among which rs749875462 in CCDC183, rs535864157 in MMP10 and rs114285050 in GPR151 showed a nominal association with ET. However, we found no significant enrichment of rare variants within these genes in cases compared with controls. Interestingly, MMP10 protein is involved in the inflammatory response to neuronal damage and has been previously associated with other neurological disorders. CONCLUSIONS We prioritized a set of promising genes, especially MMP10, for further genetic and functional studies in ET. Our study suggests that rare deleterious coding variants that markedly increase susceptibility to ET are likely to be found in many genes. Future studies are needed to replicate and further infer biological mechanisms and potential disease causality for our identified genes.
Collapse
Affiliation(s)
- Monica Diez-Fairen
- Fundació Docència i Recerca MútuaTerrassa, Movement Disorders Unit, Department of Neurology, University Hospital Mútua Terrassa, Terrassa, Barcelona, Spain
| | - Gabrielle Houle
- Department of Human Genetics, McGill University, Montréal, Quebec, Canada; Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Sara Ortega-Cubero
- Department of Neurology and Neurosurgery, Hospital Universitario de Burgos, Burgos, Spain
| | - Sara Bandres-Ciga
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA), Granada, Spain
| | - Ignacio Alvarez
- Fundació Docència i Recerca MútuaTerrassa, Movement Disorders Unit, Department of Neurology, University Hospital Mútua Terrassa, Terrassa, Barcelona, Spain
| | - Maria Carcel
- Fundació Docència i Recerca MútuaTerrassa, Movement Disorders Unit, Department of Neurology, University Hospital Mútua Terrassa, Terrassa, Barcelona, Spain
| | - Laura Ibañez
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Maria Victoria Fernandez
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - John P Budde
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Jean-Rémi Trotta
- Centre Nacional d'Anàlisis Genòmic (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain & Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Raúl Tonda
- Centre Nacional d'Anàlisis Genòmic (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain & Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jessica X Chong
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Michael J Bamshad
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA; Seattle Children's Hospital, Seattle, WA, 98105, USA; Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | | | - Miquel Aguilar
- Fundació Docència i Recerca MútuaTerrassa, Movement Disorders Unit, Department of Neurology, University Hospital Mútua Terrassa, Terrassa, Barcelona, Spain
| | - Juan P Tartari
- Fundació Docència i Recerca MútuaTerrassa, Movement Disorders Unit, Department of Neurology, University Hospital Mútua Terrassa, Terrassa, Barcelona, Spain
| | - Alexandre Gironell
- Movement Disorders Unit, Neurology Department, Hospital de Sant Pau and Sant Pau Biomedical Research Institute, Barcelona, 08026, Spain
| | - Elena García-Martín
- University Institute of Molecular Pathology Biomarkers, UNEx. ARADyAL Instituto de Salud Carlos III, Cáceres, Spain
| | - Jose Ag Agundez
- University Institute of Molecular Pathology Biomarkers, UNEx. ARADyAL Instituto de Salud Carlos III, Cáceres, Spain
| | | | | | - Manel Fernandez
- María de Maeztu Unit of Excellence, Institute of Neurosciences, University of Barcelona, MDM-2017-0729, Ministry of Science, Innovation and Universities, Spain; Parkinson's Disease & Movement Disorders Unit, Department of Neurology, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Francesc Valldeoriola
- Parkinson's Disease & Movement Disorders Unit, Department of Neurology, Hospital Clínic, IDIBAPS, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Maria Jose Marti
- Parkinson's Disease & Movement Disorders Unit, Department of Neurology, Hospital Clínic, IDIBAPS, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Eduard Tolosa
- Parkinson's Disease & Movement Disorders Unit, Department of Neurology, Hospital Clínic, IDIBAPS, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Francisco Coria
- Clinic for Nervous Disorders, Service of Neurology, Son Espases University Hospital, Palma de Mallorca, Spain
| | - Maria A Pastor
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Carles Vilariño-Güell
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Alex Rajput
- Saskatchewan Movement Disorders Program, University of Saskatchewan/Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
| | - Patrick A Dion
- Department of Human Genetics, McGill University, Montréal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, H3A 2B4, Quebec, Canada
| | - Carlos Cruchaga
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Guy A Rouleau
- Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, H3A 2B4, Quebec, Canada
| | - Pau Pastor
- Fundació Docència i Recerca MútuaTerrassa, Movement Disorders Unit, Department of Neurology, University Hospital Mútua Terrassa, Terrassa, Barcelona, Spain.
| |
Collapse
|
27
|
Liang D, Zhao Y, Pan H, Zhou X, He R, Zhou X, Yang J, Wang Y, Zhou X, Zhou Z, Xu Q, Yan X, Li J, Guo J, Tang B, Sun Q. Rare variant analysis of essential tremor-associated genes in early-onset Parkinson's disease. Ann Clin Transl Neurol 2020; 8:119-125. [PMID: 33185019 PMCID: PMC7818165 DOI: 10.1002/acn3.51248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 11/16/2022] Open
Abstract
Objective Parkinson’s disease (PD) and essential tremor (ET) are the two most common movement disorders. A significant overlap in clinical features, epidemiology, imaging, and pathology suggests that PD and ET may also share common genetic risk factors. Previous studies have only assessed a limited number of ET‐associated genes in PD patients and vice versa. Consequently, the genetic association between PD and ET remains incompletely characterized. In this study, we systematically investigated a potential association between rare coding variants in ET‐associated genes and PD, in a relatively large Chinese population cohort. Methods To investigate the genetic association between ET and PD, we performed the sequence kernel association testing (SKAT‐O) to explore the variant burden of 33 ET‐associated genes, using whole‐exome sequencing (WES) data from 1494 early‐onset PD (EOPD) patients and 1357 control subjects from mainland China. Results We report that rare loss‐of‐function and damaging missense variants of TNEM4 are suggestively associated with EOPD (P = 0.026), damaging missense variants of TNEM4 alone are also suggestively associated with EOPD (P = 0.032). No other rare damaging variants in ET‐related genes were significantly associated with EOPD. Interpretation This is the first systematic analysis of ET‐associated genes in EOPD. The suggestive association between TNEM4 and EOPD provides new evidence for a genetic link between ET and PD.
Collapse
Affiliation(s)
- Dongxiao Liang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yuwen Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hongxu Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xun Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Runcheng He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiaoxia Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jinxia Yang
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yige Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiaoting Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhou Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, 41008, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, 41008, China
| | - Jinchen Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, 41008, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, 41008, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, 41008, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China
| | - Qiying Sun
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, 41008, China
| |
Collapse
|
28
|
Chen X, Zhou R, Shan K, Sun Y, Yan B, Sun X, Wang J. Circular RNA Expression Profiling Identifies Glaucoma-Related Circular RNAs in Various Chronic Ocular Hypertension Rat Models. Front Genet 2020; 11:556712. [PMID: 33133146 PMCID: PMC7575816 DOI: 10.3389/fgene.2020.556712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022] Open
Abstract
Circular RNAs are characterized as a class of covalently closed circular RNA transcripts and are associated with a variety of cellular processes and neurological diseases by sponging microRNAs. Expression profiling of circular RNAs in glaucoma, which is a form of optic neuropathy, has not been performed to date. The most common characteristic of all forms of glaucoma is the loss of retinal ganglion cells. While the pathogenesis of glaucoma is not fully understood, intraocular pressure is unquestionably the only proven modifiable factor which makes chronic ocular hypertension (COH) animals the classical glaucoma models. Based on these findings, we completed the first in-depth study of rat retinal circular RNA expression profiling to identify probable biomarkers for the diagnosis of glaucoma. Two ocular hypertension models were induced by episcleral vein ligation (EVL) and microbead injection in rats. Overall, 15,819 circular RNA were detected. Furthermore, 3,502 differentially expressed circular RNAs verified in both COH rats were identified, of which 691 were upregulated and 2,811 were downregulated. Seven significantly downregulated (both log2FoldChange < -2.5 and adjusted P < 0.001) and seven significantly upregulated (both log2FoldChange > 2.5 and adjusted P < 0.001) circular RNAs were shown. Six target microRNAs aligned with the top 14 circular RNAs were identified. According to the construction of the circular RNA-microRNA network and circBase information, only RNO_CIRCpedia_1775 had the homologous hsa_circ_0023826 in the human genome. The hsa_circ_0023826 and mRNA of the host gene TENM4 (teneurin transmembrane protein 4) were validated in aqueous humor samples of five glaucoma patients and five cataract control patients. The expression of hsa_circ_0023826 showed a significant decrease in glaucoma patients, while TENM4 mRNA showed no significant difference compared to cataract patients (P = 0.024 and P = 0.294, respectively). The results of this study comprehensively characterized the expression profiles of circular RNA in glaucoma-affected eyes, as verified by two different ocular hypertension rat models. Together with the target microRNAs underlying the top differentially expressed circular RNAs, a new target of hsa_circ_0023826 and its host gene TENM4 were identified and further verified in the aqueous humor of glaucoma patients, indicating a promising biomarker for the disease.
Collapse
Affiliation(s)
- Xiaoxiao Chen
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Rongmei Zhou
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China
| | - Kun Shan
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China
| | - Yanan Sun
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China
| | - Biao Yan
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China
| | - Xinghuai Sun
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Jiajian Wang
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Rodríguez-Muñoz M, Cortés-Montero E, Garzón-Niño J, Sánchez-Blázquez P. The ALS-related σ1R E102Q Mutant Eludes Ligand Control and Exhibits Anomalous Response to Calcium. Int J Mol Sci 2020; 21:E7339. [PMID: 33020464 PMCID: PMC7582951 DOI: 10.3390/ijms21197339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 01/28/2023] Open
Abstract
Sigma receptor type 1 (σ1R) is a transmembrane protein expressed throughout the central nervous system and in certain peripheral tissues. The human σ1R E102Q mutation causes juvenile amyotrophic lateral sclerosis (ALS), likely by inducing a series of alterations in calcium efflux from the endoplasmic reticulum (ER) to mitochondria that affects calcium homeostasis and cellular survival. Here, we report the influence of calcium on σ1R E102Q associations with glutamate N-methyl-D-aspartate receptors (NMDARs), binding immunoglobulin protein (BiP), and transient receptor potential calcium channels A1, V1, and M8. The mutant protein inhibited the binding of calmodulin to these calcium channels and interacted less with BiP than wild-type σ1R, thereby contributing to calcium homeostasis dysfunction. Mutant σ1R, but not wild-type σ1R, strongly bound to histidine triad nucleotide binding protein 1, which regulates neuromuscular synaptic organization and target selection through teneurin 1. While ligands regulated the association of σ1R wild-type with NMDARs and BiP, they failed to modulate the interaction between these proteins and the σ1R E102Q mutant. Thus, the σ1R E102Q mutant exhibited an anomalous response to cytosolic calcium levels, altered affinity for target proteins, and a loss of response to regulatory ligands. We believe that these modifications may contribute to the onset of juvenile ALS.
Collapse
Affiliation(s)
| | | | | | - Pilar Sánchez-Blázquez
- Neuropharmacology, Cajal Institute, CSIC, Avenida Doctor Arce, 37. 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (J.G.-N.)
| |
Collapse
|
30
|
Ma X, Huang M, Zheng M, Dai C, Song Q, Zhang Q, Li Q, Gu X, Chen H, Jiang G, Yu Y, Liu X, Li S, Wang G, Chen H, Lu L, Gao X. ADSCs-derived extracellular vesicles alleviate neuronal damage, promote neurogenesis and rescue memory loss in mice with Alzheimer's disease. J Control Release 2020; 327:688-702. [PMID: 32931898 DOI: 10.1016/j.jconrel.2020.09.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
Despite the various mechanisms that involved in the pathogenesis of Alzheimer's disease (AD), neuronal damage and synaptic dysfunction are the key events leading to cognition impairment. Therefore, neuroprotection and neurogenesis would provide essential alternatives to the rescue of AD cognitive function. Here we demonstrated that extracellular vesicles secreted from adipose-derived mesenchymal stem cells (ADSCs-derived EVs, abbreviated as EVs) entered the brain quickly and efficiently following intranasal administration, and majorly accumulated in neurons within the central nervous system (CNS). Proteomics analysis showed that EVs contained multiple proteins possessing neuroprotective and neurogenesis activities, and neuronal RNA sequencing showed genes enrichment in neuroprotection and neurogenesis following the treatment with EVs. As a result, EVs exerted powerful neuroprotective effect on Aβ1-42 oligomer or glutamate-induced neuronal toxicity, effectively ameliorated neurologic damage in the whole brain areas, remarkably increased newborn neurons and powerfully rescued memory deficits in APP/PS1 transgenic mice. EVs also reduced Aβ deposition and decreased microglia activation although in a less extent. Collectively, here we provide direct evidence that ADSCs-derived EVs may potentially serve as an alternative for AD therapy through alleviating neuronal damage and promoting neurogenesis.
Collapse
Affiliation(s)
- Xinyi Ma
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Meng Huang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mengna Zheng
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chengxiang Dai
- Cellular Biomedicine Group, Inc., Shanghai 201210, China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| | - Xiao Gu
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huan Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ye Yu
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shan Dong Road, Shanghai 200001, China
| | - Xuesong Liu
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shan Dong Road, Shanghai 200001, China
| | - Suke Li
- Cellular Biomedicine Group, Inc., Shanghai 201210, China
| | - Gang Wang
- Department of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Liangjing Lu
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shan Dong Road, Shanghai 200001, China.
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
31
|
Effects of EHP-101 on inflammation and remyelination in murine models of Multiple sclerosis. Neurobiol Dis 2020; 143:104994. [PMID: 32599064 DOI: 10.1016/j.nbd.2020.104994] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/05/2020] [Accepted: 06/20/2020] [Indexed: 01/26/2023] Open
Abstract
Multiple Sclerosis (MS) is characterized by a combination of inflammatory and neurodegenerative processes in the spinal cord and the brain. Natural and synthetic cannabinoids such as VCE-004.8 have been studied in preclinical models of MS and represent promising candidates for drug development. VCE-004.8 is a multitarget synthetic cannabidiol (CBD) derivative acting as a dual Peroxisome proliferator-activated receptor-gamma/Cannabinoid receptor type 2 (PPARγ/CB2) ligand agonist that also activates the Hypoxia-inducible factor (HIF) pathway. EHP-101 is an oral lipidic formulation of VCE-004.8 that has shown efficacy in several preclinical models of autoimmune, inflammatory, fibrotic, and neurodegenerative diseases. EHP-101 alleviated clinical symptomatology in EAE and transcriptomic analysis demonstrated that EHP-101 prevented the expression of many inflammatory genes closely associated with MS pathophysiology in the spinal cord. EHP-101 normalized the expression of several genes associated with oligodendrocyte function such as Teneurin 4 (Tenm4) and Gap junction gamma-3 (Gjc3) that were downregulated in EAE. EHP-101 treatment prevented microglia activation and demyelination in both the spinal cord and the brain. Moreover, EAE was associated with a loss in the expression of Oligodendrocyte transcription factor 2 (Olig2) in the corpus callosum, a marker for oligodendrocyte differentiation, which was restored by EHP-101 treatment. In addition, EHP-101 enhanced the expression of glutathione S-transferase pi (GSTpi), a marker for mature oligodendrocytes in the brain. We also found that a diet containing 0.2% cuprizone for six weeks induced a clear loss of myelin in the brain measured by Cryomyelin staining and Myelin basic protein (MBP) expression. Moreover, EHP-101 also prevented cuprizone-induced microglial activation, astrogliosis and reduced axonal damage. Our results provide evidence that EHP-101 showed potent anti-inflammatory activity, prevented demyelination, and enhanced remyelination. Therefore, EHP-101 represents a promising drug candidate for the potential treatment of different forms of MS.
Collapse
|
32
|
Development of type I/II oligodendrocytes regulated by teneurin-4 in the murine spinal cord. Sci Rep 2020; 10:8611. [PMID: 32451386 PMCID: PMC7248063 DOI: 10.1038/s41598-020-65485-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 04/29/2020] [Indexed: 11/08/2022] Open
Abstract
In the spinal cord, the axonal tracts with various caliber sizes are myelinated by oligodendrocytes and function as high-velocity ways for motor and sensory nerve signals. In some neurological disorders, such as multiple sclerosis, demyelination of small caliber axons is observed in the spinal cord. While type I/II oligodendrocytes among the four types are known to myelinate small diameter axons, their characteristics including identification of regulating molecules have not been understood yet. Here, we first found that in the wild-type mouse spinal cord, type I/II oligodendrocytes, positive for carbonic anhydrase II (CAII), were located in the corticospinal tract, fasciculus gracilis, and the inside part of ventral funiculus, in which small diameter axons existed. The type I/II oligodendrocytes started to appear between postnatal day (P) 7 and 11. We further analyzed the type I/II oligodendrocytes in the mutant mice, whose small diameter axons were hypomyelinated due to the deficiency of teneurin-4. In the teneurin-4 deficient mice, type I/II oligodendrocytes were significantly reduced, and the onset of the defect was at P11. Our results suggest that CAII-positive type I/II oligodendrocytes myelinate small caliber axons in the spinal cord and teneurin-4 is the responsible molecule for the generation of type I/II oligodendrocytes.
Collapse
|
33
|
Yan YP, Xu CY, Gu LY, Zhang B, Shen T, Gao T, Tian J, Pu JL, Yin XZ, Zhang BR, Zhao GH. Genetic testing of FUS, HTRA2, and TENM4 genes in Chinese patients with essential tremor. CNS Neurosci Ther 2020; 26:837-841. [PMID: 32196977 PMCID: PMC7366735 DOI: 10.1111/cns.13305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Essential tremor (ET) is one of the most prevalent movement disorders. The genetic etiology of ET has not been well defined although a significant proportion (≥50%) are familial cases. Linkage analysis and genome‐wide association studies (GWASs) have identified several risk variants. In recent years, whole‐exome sequencing of ET has revealed several specific causal variants in FUS (p.Q290X), HTRA2 (p.G399S), and TENM4 (c.4324 G>A, c.4100C>A, and c.3412G>A) genes. Objective To investigate the genetic contribution of these three genes to ET, the protein‐coding sequences of FUS, HTRA2, and TENM4 were analyzed in a total of 238 ET patients and 272 controls from eastern China using direct Sanger sequencing. Results We identified two synonymous coding single nucleotide polymorphisms (SNPs), rs741810 and rs1052352 in FUS, and three previously reported synonymous SNPs, rs11237621, rs689369, and rs2277277 in TENM4. No nonsynonymous exonic variants were identified in these subjects. We found that the frequency of the rs1052352C allele was significantly higher (P = .001) in the ET group than in the control group. Conclusion Overall, our findings suggest that rs1052352 of FUS might contribute to ET risk in Chinese population.
Collapse
Affiliation(s)
- Ya-Ping Yan
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong-Ying Xu
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lu-Yan Gu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bo Zhang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Shen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Gao
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Tian
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Li Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin-Zhen Yin
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bao-Rong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guo-Hua Zhao
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
34
|
The extracellular domain of teneurin-4 promotes cell adhesion for oligodendrocyte differentiation. Biochem Biophys Res Commun 2019; 523:171-176. [PMID: 31839217 DOI: 10.1016/j.bbrc.2019.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 12/01/2019] [Indexed: 01/06/2023]
Abstract
Cell adhesion between oligodendrocytes and neuronal axons is a critical step for myelination that enables the rapid propagation of action potential in the central nervous system. Here, we show that the transmembrane protein teneurin-4 plays a role in the cell adhesion required for the differentiation of oligodendrocytes. We found that teneurin-4 formed molecular complexes with all of the four teneurin family members and promoted cell-cell adhesion. Oligodendrocyte lineage cells attached to the recombinant extracellular domain of all the teneurins and formed well-branched cell processes. In an axon-mimicking nanofibers assay, nanofibers coated with the recombinant teneurin-4 extracellular domain increased the differentiation of oligodendrocytes. Our results show that teneurin-4 binds to all teneurins through their extracellular domain, which facilitates the oligodendrocyte-axon adhesion, and promotes oligodendrocyte differentiation via its homophilic interaction.
Collapse
|
35
|
Abstract
Essential tremor (ET) is a neurological movement disorder characterised by bilateral limb kinetic/postural tremor, with or without tremor in other body parts including head, voice and lower limbs. Since no causative genes for ET have been identified, it is likely that the disorder occurs as a result of complex genetic factors interacting with various cellular and environmental factors that can result in abnormal function of circuitry involving the cerebello-thalamo-cortical pathway. Genetic analyses have uncovered at least 14 loci and 11 genes that are related to ET, as well as various risk or protective genetic factors. Limitations in ET genetic analyses include inconsistent disease definition, small sample size, varied ethnic backgrounds and many other factors that may contribute to paucity of relevant genetic data in ET. Genetic analyses, coupled with functional and animal studies, have led to better insights into possible pathogenic mechanisms underlying ET. These genetic studies may guide the future development of genetic testing and counselling, and specific, pathogenesis-targeted, therapeutic strategies.
Collapse
|
36
|
Cadwell CR, Bhaduri A, Mostajo-Radji MA, Keefe MG, Nowakowski TJ. Development and Arealization of the Cerebral Cortex. Neuron 2019; 103:980-1004. [PMID: 31557462 PMCID: PMC9245854 DOI: 10.1016/j.neuron.2019.07.009] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 05/15/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022]
Abstract
Adult cortical areas consist of specialized cell types and circuits that support unique higher-order cognitive functions. How this regional diversity develops from an initially uniform neuroepithelium has been the subject of decades of seminal research, and emerging technologies, including single-cell transcriptomics, provide a new perspective on area-specific molecular diversity. Here, we review the early developmental processes that underlie cortical arealization, including both cortex intrinsic and extrinsic mechanisms as embodied by the protomap and protocortex hypotheses, respectively. We propose an integrated model of serial homology whereby intrinsic genetic programs and local factors establish early transcriptomic differences between excitatory neurons destined to give rise to broad "proto-regions," and activity-dependent mechanisms lead to progressive refinement and formation of sharp boundaries between functional areas. Finally, we explore the potential of these basic developmental processes to inform our understanding of the emergence of functional neural networks and circuit abnormalities in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Cathryn R Cadwell
- Department of Anatomic Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Aparna Bhaduri
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94122, USA; The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research at the University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mohammed A Mostajo-Radji
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94122, USA; The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research at the University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew G Keefe
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tomasz J Nowakowski
- The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research at the University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
37
|
Burbach JPH, Meijer DH. Latrophilin's Social Protein Network. Front Neurosci 2019; 13:643. [PMID: 31297045 PMCID: PMC6608557 DOI: 10.3389/fnins.2019.00643] [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: 03/21/2019] [Accepted: 06/05/2019] [Indexed: 01/06/2023] Open
Abstract
Latrophilins (LPHNs) are adhesion GPCRs that are originally discovered as spider's toxin receptors, but are now known to be involved in brain development and linked to several neuronal and non-neuronal disorders. Latrophilins act in conjunction with other cell adhesion molecules and may play a leading role in its network organization. Here, we focus on the main protein partners of latrophilins, namely teneurins, FLRTs and contactins and summarize their respective temporal and spatial expression patterns, links to neurodevelopmental disorders as well as their structural characteristics. We discuss how more recent insights into the separate cell biological functions of these proteins shed light on the central role of latrophilins in this network. We postulate that latrophilins control the refinement of synaptic properties of specific subtypes of neurons, requiring discrete combinations of proteins.
Collapse
Affiliation(s)
- J Peter H Burbach
- Department of Translational Neuroscience, UMCU Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Dimphna H Meijer
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
38
|
Moreno-Salinas AL, Avila-Zozaya M, Ugalde-Silva P, Hernández-Guzmán DA, Missirlis F, Boucard AA. Latrophilins: A Neuro-Centric View of an Evolutionary Conserved Adhesion G Protein-Coupled Receptor Subfamily. Front Neurosci 2019; 13:700. [PMID: 31354411 PMCID: PMC6629964 DOI: 10.3389/fnins.2019.00700] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
The adhesion G protein-coupled receptors latrophilins have been in the limelight for more than 20 years since their discovery as calcium-independent receptors for α-latrotoxin, a spider venom toxin with potent activity directed at neurotransmitter release from a variety of synapse types. Latrophilins are highly expressed in the nervous system. Although a substantial amount of studies has been conducted to describe the role of latrophilins in the toxin-mediated action, the recent identification of endogenous ligands for these receptors helped confirm their function as mediators of adhesion events. Here we hypothesize a role for latrophilins in inter-neuronal contacts and the formation of neuronal networks and we review the most recent information on their role in neurons. We explore molecular, cellular and behavioral aspects related to latrophilin adhesion function in mice, zebrafish, Drosophila melanogaster and Caenorhabditis elegans, in physiological and pathophysiological conditions, including autism spectrum, bipolar, attention deficit and hyperactivity and substance use disorders.
Collapse
Affiliation(s)
- Ana L. Moreno-Salinas
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Monserrat Avila-Zozaya
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Paul Ugalde-Silva
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - David A. Hernández-Guzmán
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Fanis Missirlis
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Antony A. Boucard
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
39
|
Leamey CA, Sawatari A. Teneurins: Mediators of Complex Neural Circuit Assembly in Mammals. Front Neurosci 2019; 13:580. [PMID: 31231187 PMCID: PMC6560073 DOI: 10.3389/fnins.2019.00580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/22/2019] [Indexed: 01/27/2023] Open
Abstract
The teneurins (Ten-m/Odz) are a family of evolutionarily ancient transmembrane molecules whose complex and multi-faceted roles in the generation of mammalian neural circuits are only beginning to be appreciated. In mammals there are four family members (Ten-m1-4). Initial expression studies in vertebrates revealed intriguing expression patterns in interconnected populations of neurons. These observations, together with biochemical and over-expression studies, led to the hypothesis that homophilic interactions between teneurins on afferent and target cells may help to guide the assembly of neural circuits. This review will focus on insights gained on teneurin function in vivo in mammals using mouse knockout models. These studies provide support for the hypothesis that homophilic interactions between teneurin molecules can guide the formation of neural connections with largely consistent results obtained in hippocampal and striatal circuits. Mapping changes obtained in the mouse visual pathway, however, suggest additional roles for these glycoproteins in the formation and specification of circuits which subserve binocular vision.
Collapse
Affiliation(s)
- Catherine A Leamey
- Discipline of Physiology, School of Medical Sciences and Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Atomu Sawatari
- Discipline of Physiology, School of Medical Sciences and Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
40
|
Hayashi C, Suzuki N. Heterogeneity of Oligodendrocytes and Their Precursor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:53-62. [PMID: 31760638 DOI: 10.1007/978-981-32-9636-7_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
While oligodendrocytes have been thought to be homogenous, a number of reports have indicated evidences of the heterogeneity of oligodendrocytes and their precursor cells, OPCs. Almost a century ago, Del Río Hortega found three and four types of oligodendrocytes with regions where they exist and their morphologies, respectively. Interfascicular oligodendrocytes are one of the three regional dependent types and are the most typical oligodendendroglial cells that myelinate axonal fibers in the white matter tracts. In the other two, perineuronal oligodendrocyes function as reserve cells for remyelination and regulate neuronal excitability, whereas perivascular oligodendrocytes may play a role in metabolic support of axons. Among the four morphological categories, type I and II oligodendrocytes form many myelin sheaths on small-diameter axons and specific signal is required for the myelination of small-diameter axons. Type III and IV oligodendrocytes myelinate a few number of axons/or one axon, whose diameters are large. A recent comprehensive gene expression analysis with single-cell RNA sequencing identifies six different populations in mature oligodendrocytes and only one population in OPCs. However, OPCs are not uniformed developmentally and regionally. Further, the capacity of OPC differentiation depends on the environments and conditions of the tissues. Taken together, oligodendrocytes and OPCs are diverse as the other cell types in the CNS. The orchestration of these cells with their specialized functions is critical for proper functioning of the CNS.
Collapse
Affiliation(s)
- Chikako Hayashi
- Department of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan
| | - Nobuharu Suzuki
- Department of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan. .,Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan.
| |
Collapse
|
41
|
Tucker RP. Teneurins: Domain Architecture, Evolutionary Origins, and Patterns of Expression. Front Neurosci 2018; 12:938. [PMID: 30618567 PMCID: PMC6297184 DOI: 10.3389/fnins.2018.00938] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/28/2018] [Indexed: 12/24/2022] Open
Abstract
Disruption of teneurin expression results in abnormal neural networks, but just how teneurins support the development of the central nervous system remains an area of active research. This review summarizes some of what we know about the functions of the various domains of teneurins, the possible evolution of teneurins from a bacterial toxin, and the intriguing patterns of teneurin expression. Teneurins are a family of type-2 transmembrane proteins. The N-terminal intracellular domain can be processed and localized to the nucleus, but the significance of this nuclear localization is unknown. The extracellular domain of teneurins is largely composed of tyrosine-aspartic acid repeats that fold into a hollow barrel, and the C-terminal domains of teneurins are stuffed, and least partly, into the barrel. A 6-bladed beta-propeller is found at the other end of the barrel. The same arrangement-6-bladed beta-propeller, tyrosine-aspartic acid repeat barrel, and the C-terminal domain inside the barrel-is seen in toxic proteins from bacteria, and there is evidence that teneurins may have evolved from a gene encoding a prokaryotic toxin via horizontal gene transfer into an ancestral choanoflagellate. Patterns of teneurin expression are often, but not always, complementary. In the central nervous system, where teneurins are best studied, interconnected populations of neurons often express the same teneurin. For example, in the chicken embryo neurons forming the tectofugal pathway express teneurin-1, whereas neurons forming the thalamofugal pathway express teneurin-2. In Drosophila melanogaster, Caenorhabditis elegans, zebrafish and mice, misexpression or knocking out teneurin expression leads to abnormal connections in the neural networks that normally express the relevant teneurin. Teneurins are also expressed in non-neuronal tissue during development, and in at least some regions the patterns of non-neuronal expression are also complementary. The function of teneurins outside the nervous system remains unclear.
Collapse
Affiliation(s)
- Richard P. Tucker
- Department of Cell Biology and Human Anatomy, University of California at Davis, Davis, CA, United States
| |
Collapse
|
42
|
Rebolledo-Jaramillo B, Ziegler A. Teneurins: An Integrative Molecular, Functional, and Biomedical Overview of Their Role in Cancer. Front Neurosci 2018; 12:937. [PMID: 30618566 PMCID: PMC6297388 DOI: 10.3389/fnins.2018.00937] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Teneurins are large transmembrane proteins originally identified in Drosophila. Their essential role in development of the central nervous system is conserved throughout species, and evidence supports their involvement in organogenesis of additional tissues. Homophilic and heterophilic interactions between Teneurin paralogues mediate cellular adhesion in crucial processes such as neuronal pathfinding and synaptic organization. At the molecular level, Teneurins are proteolytically processed into distinct subdomains that have been implicated in extracellular and intracellular signaling, and in transcriptional regulation. Phylogenetic studies have shown a high degree of intra- and interspecies conservation of Teneurin genes. Accordingly, the occurrence of genetic variants has been associated with functional and phenotypic alterations in experimental systems, and with some inherited or sporadic conditions. Recently, tumor-related variations in Teneurin gene expression have been associated with patient survival in different cancers. Although these findings were incidental and molecular mechanisms were not addressed, they suggested a potential utility of Teneurin transcript levels as biomarkers for disease prognosis. Mutations and chromosomal alterations affecting Teneurin genes have been found occasionally in tumors, but literature remains scarce. The analysis of open-access molecular and clinical datasets derived from large oncologic cohorts provides an invaluable resource for the identification of additional somatic mutations. However, Teneurin variants have not been classified in terms of pathogenic risk and their phenotypic impact remains unknown. On this basis, is it plausible to hypothesize that Teneurins play a role in carcinogenesis? Does current evidence support a tumor suppressive or rather oncogenic function for these proteins? Here, we comprehensively discuss available literature with integration of molecular evidence retrieved from open-access databases. We show that Teneurins undergo somatic changes comparable to those of well-established cancer genes, and discuss their involvement in cancer-related signaling pathways. Current data strongly suggest a functional contribution of Teneurins to human carcinogenesis.
Collapse
Affiliation(s)
| | - Annemarie Ziegler
- Center for Genetics and Genomics, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
43
|
Juttukonda MR, Franco G, Englot DJ, Lin YC, Petersen KJ, Trujillo P, Hedera P, Landman BA, Kang H, Donahue MJ, Konrad PE, Dawant BM, Claassen DO. White matter differences between essential tremor and Parkinson disease. Neurology 2018; 92:e30-e39. [PMID: 30504432 DOI: 10.1212/wnl.0000000000006694] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/05/2018] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess white matter integrity in patients with essential tremor (ET) and Parkinson disease (PD) with moderate to severe motor impairment. METHODS Sedated participants with ET (n = 57) or PD (n = 99) underwent diffusion tensor imaging (DTI) and fractional anisotropy, mean diffusivity, axial diffusivity, and radial diffusivity values were computed. White matter tracts were defined using 3 well-described atlases. To determine candidate white matter regions that differ between ET and PD groups, a bootstrapping analysis was applied using the least absolute shrinkage and selection operator. Linear regression was applied to assess magnitude and direction of differences in DTI metrics between ET and PD populations in the candidate regions. RESULTS Fractional anisotropy values that differentiate ET from PD localize primarily to thalamic and visual-related pathways, while diffusivity differences localized to the cerebellar peduncles. Patients with ET exhibited lower fractional anisotropy values than patients with PD in the lateral geniculate body (p < 0.01), sagittal stratum (p = 0.01), forceps major (p = 0.02), pontine crossing tract (p = 0.03), and retrolenticular internal capsule (p = 0.04). Patients with ET exhibited greater radial diffusivity values than patients with PD in the superior cerebellar peduncle (p < 0.01), middle cerebellar peduncle (p = 0.05), and inferior cerebellar peduncle (p = 0.05). CONCLUSIONS Regionally, distinctive white matter microstructural values in patients with ET localize to the cerebellar peduncles and thalamo-cortical visual pathways. These findings complement recent functional imaging studies in ET but also extend our understanding of putative physiologic features that account for distinctions between ET and PD.
Collapse
Affiliation(s)
- Meher R Juttukonda
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Giulia Franco
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Dario J Englot
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Ya-Chen Lin
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Kalen J Petersen
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Paula Trujillo
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Peter Hedera
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Bennett A Landman
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Hakmook Kang
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Manus J Donahue
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Peter E Konrad
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Benoit M Dawant
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN
| | - Daniel O Claassen
- From the Departments of Radiology and Radiological Sciences (M.R.J., M.J.D.), Neurological Surgery (D.J.E., P.E.K.), Biostatistics (Y.-C.L., H.K.), Neurology (P.T., P.H., M.J.D.), and Psychiatry (M.J.D.), Vanderbilt University Medical Center, Nashville, TN; Department of Pathophysiology and Transplantation (G.F.) University of Milan, Italy; and Chemical and Physical Biology Program (K.J.P.) and Departments of Electrical Engineering (B.A.L., B.M.D.), Computer Engineering (B.A.L., B.M.D.), Computer Science and Biomedical Engineering (B.A.L., B.M.D.), and Neurology (D.O.C.), Vanderbilt University, Nashville, TN.
| |
Collapse
|
44
|
Südhof TC. Towards an Understanding of Synapse Formation. Neuron 2018; 100:276-293. [PMID: 30359597 PMCID: PMC6226307 DOI: 10.1016/j.neuron.2018.09.040] [Citation(s) in RCA: 379] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 09/10/2018] [Accepted: 09/25/2018] [Indexed: 12/17/2022]
Abstract
Synapses are intercellular junctions specialized for fast, point-to-point information transfer from a presynaptic neuron to a postsynaptic cell. At a synapse, a presynaptic terminal secretes neurotransmitters via a canonical release machinery, while a postsynaptic specialization senses neurotransmitters via diverse receptors. Synaptic junctions are likely organized by trans-synaptic cell-adhesion molecules (CAMs) that bidirectionally orchestrate synapse formation, restructuring, and elimination. Many candidate synaptic CAMs were described, but which CAMs are central actors and which are bystanders remains unclear. Moreover, multiple genes encoding synaptic CAMs were linked to neuropsychiatric disorders, but the mechanisms involved are unresolved. Here, I propose that engagement of multifarious synaptic CAMs produces parallel trans-synaptic signals that mediate the establishment, organization, and plasticity of synapses, thereby controlling information processing by neural circuits. Among others, this hypothesis implies that synapse formation can be understood in terms of inter- and intracellular signaling, and that neuropsychiatric disorders involve an impairment in such signaling.
Collapse
Affiliation(s)
- Thomas C Südhof
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
45
|
Differentiation of Oligodendrocyte Precursor Cells from Sox10-Venus Mice to Oligodendrocytes and Astrocytes. Sci Rep 2017; 7:14133. [PMID: 29074959 PMCID: PMC5658394 DOI: 10.1038/s41598-017-14207-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022] Open
Abstract
Oligodendrocytes are well known as myelin-forming cells in the central nervous system (CNS). However, detailed mechanisms of oligodendrocyte differentiation and myelination are poorly understood, particularly due to the difficulty of the purification of murine oligodendrocyte precursor cells (OPCs). We have recently established a transgenic mouse line that expresses a fluorescent protein Venus under the promoter of Sox10, whose expression is restricted to OPCs and oligodendrocytes in the CNS. Here, we have characterized Venus-positive cells from the Sox10-Venus mouse brain for analyzing oligodendrocyte differentiation. We first purified Venus-positive cells from the postnatal day 0-2 brain by flow cytometry. Most of the Venus-positive cells expressed NG2, an OPC marker. After induction of differentiation, an increased population of galactocerebroside-positive oligodendrocytes and decrease of OPCs were observed in the Venus-positive culture. Furthermore, a time-lapse analysis showed that Venus-positive oligodendrocytes dynamically changed their morphology with highly branched cell processes during differentiation. In addition, we found that Venus-positive OPCs were able to differentiate to type II astrocytes. In vivo, OPCs and oligodendrocytes express Venus, and some of astrocytes were positive for Venus in the ventral cortex. Taken together, the Sox10-Venus mouse system is useful for analyzing differentiation and multipotency of murine OPCs.
Collapse
|
46
|
Graumann R, Di Capua GA, Oyarzún JE, Vásquez MA, Liao C, Brañes JA, Roa I, Casanello P, Corvalán AH, Owen GI, Delgado I, Zangemeister-Wittke U, Ziegler A. Expression of teneurins is associated with tumor differentiation and patient survival in ovarian cancer. PLoS One 2017; 12:e0177244. [PMID: 28472127 PMCID: PMC5417686 DOI: 10.1371/journal.pone.0177244] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/24/2017] [Indexed: 11/29/2022] Open
Abstract
Teneurins are a family of highly conserved pair-rule proteins involved in morphogenesis and development of the central nervous system. Their function in adult tissues and in disease is largely unknown. Recent evidence suggests a role for dysregulated expression of Teneurins in human tumors, but systematic investigations are missing. Here, we investigated Teneurin-2 and Teneurin-4 expression in various cancer cell lines and in ovarian tumor tissues. Teneurin-2 and Teneurin-4 were expressed in most of the breast cancer cell lines tested. Teneurin-4 was also detected in ovarian cancer cell lines, and throughout ovarian tumors and normal ovary tissue. Ovarian tumors with low Teneurin-4 expression showed less differentiated phenotypes and these patients had shorter mean overall survival. Similarly, Teneurin-2 expression correlated with overall survival as well, especially in patients with serous tumors. In the various cell lines, 5-Aza-cytidine-induced changes in DNA methylation did not alter expression of Teneurin-2 and Teneurin-4, despite the existence of predicted CpG islands in both genes. Interestingly, however, we found evidence for the control of Teneurin-2 expression by the oncogenic growth factor FGF8. Furthermore, we identified multiple transcript splicing variants for Teneurin-2 and Teneurin-4, indicating complex gene expression patterns in malignant cells. Finally, downregulation of Teneurin-4 expression using siRNA caused a cell-type dependent increase in proliferation and resistance to cisplatin. Altogether, our data suggest that low Teneurin-4 expression provides a growth advantage to cancer cells and marks an undifferentiated state characterized by increased drug resistance and clinical aggressiveness. We conclude that Teneurin-2 and Teneurin-4 expression levels could be of prognostic value in ovarian cancer.
Collapse
Affiliation(s)
- Rebecca Graumann
- Center for Genetics and Genomics, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Gabriella A. Di Capua
- Center for Genetics and Genomics, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Juan E. Oyarzún
- Center for Genetics and Genomics, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Marcos A. Vásquez
- Center for Genetics and Genomics, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Christine Liao
- Center for Genetics and Genomics, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Jorge A. Brañes
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Iván Roa
- Division of Pathology, Clínica Alemana de Santiago, Santiago, Chile
| | - Paola Casanello
- Perinatology Research Laboratory, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro H. Corvalán
- Advanced Center for Chronic Diseases (ACCDiS), and UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gareth I. Owen
- Advanced Center for Chronic Diseases (ACCDiS), and UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Iris Delgado
- Center for Epidemiology and Health Policies, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | | | - Annemarie Ziegler
- Center for Genetics and Genomics, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
- * E-mail:
| |
Collapse
|
47
|
Crowe EP, Tuzer F, Gregory BD, Donahue G, Gosai SJ, Cohen J, Leung YY, Yetkin E, Nativio R, Wang LS, Sell C, Bonini NM, Berger SL, Johnson FB, Torres C. Changes in the Transcriptome of Human Astrocytes Accompanying Oxidative Stress-Induced Senescence. Front Aging Neurosci 2016; 8:208. [PMID: 27630559 PMCID: PMC5005348 DOI: 10.3389/fnagi.2016.00208] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/15/2016] [Indexed: 01/23/2023] Open
Abstract
Aging is a major risk factor for many neurodegenerative disorders. A key feature of aging biology that may underlie these diseases is cellular senescence. Senescent cells accumulate in tissues with age, undergo widespread changes in gene expression, and typically demonstrate altered, pro-inflammatory profiles. Astrocyte senescence has been implicated in neurodegenerative disease, and to better understand senescence-associated changes in astrocytes, we investigated changes in their transcriptome using RNA sequencing. Senescence was induced in human fetal astrocytes by transient oxidative stress. Brain-expressed genes, including those involved in neuronal development and differentiation, were downregulated in senescent astrocytes. Remarkably, several genes indicative of astrocytic responses to injury were also downregulated, including glial fibrillary acidic protein and genes involved in the processing and presentation of antigens by major histocompatibility complex class II proteins, while pro-inflammatory genes were upregulated. Overall, our findings suggest that senescence-related changes in the function of astrocytes may impact the pathogenesis of age-related brain disorders.
Collapse
Affiliation(s)
- Elizabeth P Crowe
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Ferit Tuzer
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Brian D Gregory
- Department of Biology, Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia PA, USA
| | - Greg Donahue
- Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Sager J Gosai
- Department of Biology, Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia PA, USA
| | - Justin Cohen
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Yuk Y Leung
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Emre Yetkin
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Raffaella Nativio
- Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Christian Sell
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| | - Nancy M Bonini
- Department of Biology, Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia PA, USA
| | - Shelley L Berger
- Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Claudio Torres
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia PA, USA
| |
Collapse
|
48
|
Benner B, Martorell AJ, Mahadevan P, Najm FJ, Tesar PJ, Freundt EC. Depletion of Olig2 in oligodendrocyte progenitor cells infected by Theiler's murine encephalomyelitis virus. J Neurovirol 2015; 22:336-48. [PMID: 26631080 DOI: 10.1007/s13365-015-0402-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 10/19/2015] [Accepted: 10/27/2015] [Indexed: 01/03/2023]
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infects the central nervous system of mice and causes a demyelinating disease that is a model for multiple sclerosis. During the chronic phase of the disease, TMEV persists in oligodendrocytes and macrophages. Lack of remyelination has been attributed to insufficient proliferation and differentiation of oligodendrocyte progenitor cells (OPCs), but the molecular mechanisms remain unknown. Here, we employed pluripotent stem cell technologies to generate pure populations of mouse OPCs to study the temporal and molecular effects of TMEV infection. Global transcriptome analysis of RNA sequencing data revealed that TMEV infection of OPCs caused significant up-regulation of 1926 genes, whereas 1853 genes were significantly down-regulated compared to uninfected cells. Pathway analysis revealed that TMEV disrupted many genes required for OPC growth and maturation. Down-regulation of Olig2, a transcription factor necessary for OPC proliferation, was confirmed by real-time PCR, immunofluorescence microscopy, and western blot analysis. Depletion of Olig2 was not found to be specific to viral strain and did not require expression of the leader (L) protein, which is a multifunctional protein important for persistence, modulation of gene expression, and cell death. These data suggest that direct infection of OPCs by TMEV may inhibit remyelination during the chronic phase of TMEV-induced demyelinating disease.
Collapse
Affiliation(s)
- Bayleigh Benner
- Department of Biology, The University of Tampa, Tampa, FL, USA
| | | | | | - Fadi J Najm
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Eric C Freundt
- Department of Biology, The University of Tampa, Tampa, FL, USA.
| |
Collapse
|
49
|
Hor H, Francescatto L, Bartesaghi L, Ortega-Cubero S, Kousi M, Lorenzo-Betancor O, Jiménez-Jiménez FJ, Gironell A, Clarimón J, Drechsel O, Agúndez JAG, Kenzelmann Broz D, Chiquet-Ehrismann R, Lleó A, Coria F, García-Martin E, Alonso-Navarro H, Martí MJ, Kulisevsky J, Hor CN, Ossowski S, Chrast R, Katsanis N, Pastor P, Estivill X. Missense mutations in TENM4, a regulator of axon guidance and central myelination, cause essential tremor. Hum Mol Genet 2015; 24:5677-86. [PMID: 26188006 DOI: 10.1093/hmg/ddv281] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/13/2015] [Indexed: 12/16/2022] Open
Abstract
Essential tremor (ET) is a common movement disorder with an estimated prevalence of 5% of the population aged over 65 years. In spite of intensive efforts, the genetic architecture of ET remains unknown. We used a combination of whole-exome sequencing and targeted resequencing in three ET families. In vitro and in vivo experiments in oligodendrocyte precursor cells and zebrafish were performed to test our findings. Whole-exome sequencing revealed a missense mutation in TENM4 segregating in an autosomal-dominant fashion in an ET family. Subsequent targeted resequencing of TENM4 led to the discovery of two novel missense mutations. Not only did these two mutations segregate with ET in two additional families, but we also observed significant over transmission of pathogenic TENM4 alleles across the three families. Consistent with a dominant mode of inheritance, in vitro analysis in oligodendrocyte precursor cells showed that mutant proteins mislocalize. Finally, expression of human mRNA harboring any of three patient mutations in zebrafish embryos induced defects in axon guidance, confirming a dominant-negative mode of action for these mutations. Our genetic and functional data, which is corroborated by the existence of a Tenm4 knockout mouse displaying an ET phenotype, implicates TENM4 in ET. Together with previous studies of TENM4 in model organisms, our studies intimate that processes regulating myelination in the central nervous system and axon guidance might be significant contributors to the genetic burden of this disorder.
Collapse
Affiliation(s)
- Hyun Hor
- Bioinformatics and Genomics Program, Centre for Genomic Regulation (CRG), Barcelona, Spain, Universitat Pompeu Fabra (UPF), Barcelona, Spain, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain, CRG CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Catalonia 08003, Spain,
| | - Ludmila Francescatto
- Center for Human Disease Modeling, Duke University, Duke University Medical Center, Durham NC 27710, USA
| | - Luca Bartesaghi
- Department of Medical Genetics, University of Lausanne, Lausanne 1005, Switzerland, Department of Neuroscience and Department of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Sara Ortega-Cubero
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research (CIMA), and Department of Neurology, Clínica Universidad de Navarra, University of Navarra School of Medicine and Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Pamplona, Navarra 31008, Spain
| | - Maria Kousi
- Center for Human Disease Modeling, Duke University, Duke University Medical Center, Durham NC 27710, USA
| | - Oswaldo Lorenzo-Betancor
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research (CIMA), and Department of Neurology, Clínica Universidad de Navarra, University of Navarra School of Medicine and Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Pamplona, Navarra 31008, Spain
| | - Felix J Jiménez-Jiménez
- Section of Neurology, Hospital Universitario del Sureste, Arganda del Rey, Madrid 28030, Spain
| | - Alexandre Gironell
- Movement Disorders Unit, Neurology Department, Hospital de Sant Pau, Barcelona, Spain, Sant Pau Biomedical Research Institute, Barcelona, Spain
| | - Jordi Clarimón
- Sant Pau Biomedical Research Institute, Barcelona, Spain, Universitat Autònoma de Barcelona and CIBERNED, Barcelona, Catalonia 08026, Spain
| | - Oliver Drechsel
- Bioinformatics and Genomics Program, Centre for Genomic Regulation (CRG), Barcelona, Spain, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | - Daniela Kenzelmann Broz
- Faculty of Sciences and Department of Biomedicine, Friedrich Miescher Institute of Biomedical Research, Novartis Research Foundation and University of Basel, Basel 4058, Switzerland
| | - Ruth Chiquet-Ehrismann
- Faculty of Sciences and Department of Biomedicine, Friedrich Miescher Institute of Biomedical Research, Novartis Research Foundation and University of Basel, Basel 4058, Switzerland
| | - Alberto Lleó
- Sant Pau Biomedical Research Institute, Barcelona, Spain
| | - Francisco Coria
- Clinic for Nervous Disorders, Service of Neurology, Son Espases University Hospital, Palma de Mallorca 07120, Spain
| | - Elena García-Martin
- Department of Biochemistry and Molecular Biology, University of Extremadura, Cáceres 10071, Spain
| | | | - Maria J Martí
- Movement Disorders Unit, Neurology Service, Hospital Clinic, CIBERNED and Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia 08036, Spain and
| | - Jaume Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de Sant Pau, Barcelona, Spain, Universitat Autònoma de Barcelona and CIBERNED, Barcelona, Catalonia 08026, Spain
| | - Charlotte N Hor
- Bioinformatics and Genomics Program, Centre for Genomic Regulation (CRG), Barcelona, Spain, Universitat Pompeu Fabra (UPF), Barcelona, Spain, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain, CRG CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Catalonia 08003, Spain
| | - Stephan Ossowski
- Bioinformatics and Genomics Program, Centre for Genomic Regulation (CRG), Barcelona, Spain, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Roman Chrast
- Department of Medical Genetics, University of Lausanne, Lausanne 1005, Switzerland, Department of Neuroscience and Department of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University, Duke University Medical Center, Durham NC 27710, USA
| | - Pau Pastor
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research (CIMA), and Department of Neurology, Clínica Universidad de Navarra, University of Navarra School of Medicine and Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Pamplona, Navarra 31008, Spain,
| | - Xavier Estivill
- Bioinformatics and Genomics Program, Centre for Genomic Regulation (CRG), Barcelona, Spain, Universitat Pompeu Fabra (UPF), Barcelona, Spain, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain, CRG CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Catalonia 08003, Spain, Dexeus Women's Health, University Hospital Quiron-Dexeus, Barcelona, Catalonia 08028, Spain
| |
Collapse
|
50
|
Ishii K, Suzuki N, Mabuchi Y, Ito N, Kikura N, Fukada S, Okano H, Takeda S, Akazawa C. Muscle Satellite Cell Protein Teneurin-4 Regulates Differentiation During Muscle Regeneration. Stem Cells 2015; 33:3017-27. [PMID: 26013034 PMCID: PMC4744701 DOI: 10.1002/stem.2058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/30/2015] [Indexed: 02/06/2023]
Abstract
Satellite cells are maintained in an undifferentiated quiescent state, but during muscle regeneration they acquire an activated stage, and initiate to proliferate and differentiate as myoblasts. The transmembrane protein teneurin-4 (Ten-4) is specifically expressed in the quiescent satellite cells; however, its cellular and molecular functions remain unknown. We therefore aimed to elucidate the function of Ten-4 in muscle satellite cells. In the tibialis anterior (TA) muscle of Ten-4-deficient mice, the number and the size of myofibers, as well as the population of satellite cells, were reduced with/without induction of muscle regeneration. Furthermore, we found an accelerated activation of satellite cells in the regenerated Ten-4-deficient TA muscle. The cell culture analysis using primary satellite cells showed that Ten-4 suppressed the progression of myogenic differentiation. Together, our findings revealed that Ten-4 functions as a crucial player in maintaining the quiescence of muscle satellite cells.
Collapse
Affiliation(s)
- Kana Ishii
- Department of Biochemistry and BiophysicsGraduate School of Health Care Sciences, Tokyo Medical and Dental UniversityTokyoJapan
| | - Nobuharu Suzuki
- Department of Biochemistry and BiophysicsGraduate School of Health Care Sciences, Tokyo Medical and Dental UniversityTokyoJapan
| | - Yo Mabuchi
- Department of Biochemistry and BiophysicsGraduate School of Health Care Sciences, Tokyo Medical and Dental UniversityTokyoJapan
| | - Naoki Ito
- Department of Molecular TherapyNational Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaTokyoJapan
| | - Naomi Kikura
- Department of Biochemistry and BiophysicsGraduate School of Health Care Sciences, Tokyo Medical and Dental UniversityTokyoJapan
| | - So‐ichiro Fukada
- Laboratory of Molecular and Cellular PhysiologyGraduate School of Pharmaceutical Sciences, Osaka UniversitySuitaOsakaJapan
| | - Hideyuki Okano
- Department of PhysiologyKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Shin'ichi Takeda
- Department of Molecular TherapyNational Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaTokyoJapan
| | - Chihiro Akazawa
- Department of Biochemistry and BiophysicsGraduate School of Health Care Sciences, Tokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|