1
|
Mair DB, Elmasli C, Kim JH, Barreto AD, Ding S, Gu L, Weinberg SH, Kim T, Kim DH, Li R. The Arp2/3 complex enhances cell migration on elastic substrates. Mol Biol Cell 2023; 34:ar67. [PMID: 36989030 PMCID: PMC10295479 DOI: 10.1091/mbc.e22-06-0243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/23/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
Cell migration on soft surfaces occurs in both physiological and pathological processes such as corticogenesis during embryonic development and cancer invasion and metastasis. The Arp2/3 complex in neural progenitor cells was previously demonstrated to be necessary for cell migration on soft elastic substrate but not on stiff surfaces, but the underlying mechanism was unclear. Here, we integrate computational and experimental approaches to elucidate how the Arp2/3 complex enables cell migration on soft surfaces. We found that lamellipodia comprised of a branched actin network nucleated by the Arp2/3 complex distribute forces over a wider area, thus decreasing stress in the substrate. Additionally, we found that interactions between parallel focal adhesions within lamellipodia prolong cell-substrate interactions by compensating for the failure of neighboring adhesions. Together with decreased substrate stress, this leads to the observed improvements in migratory ability on soft substrates in cells utilizing lamellipodia-dependent mesenchymal migration when compared with filopodia-based migration. These results show that the Arp2/3 complex-dependent lamellipodia provide multiple distinct mechanical advantages to gliomas migrating on soft 2D substrates, which can contribute to their invasive potential.
Collapse
Affiliation(s)
- Devin B. Mair
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ceylin Elmasli
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218
| | - June Hyung Kim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907
| | - Amanda D. Barreto
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Biomedical Engineering, Florida International University College of Engineering and Computing, Miami, FL 33199
| | - Supeng Ding
- Department of Materials Science and Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21205
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21205
| | - Seth H. Weinberg
- Department of Biomedical Engineering, The Ohio State University, Wexner Medical Center, Columbus, OH 43210
- Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, Columbus, OH 43210
| | - Taeyoon Kim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Rong Li
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218
- Mechanobiology Institute and Department of Biological Science, National University of Singapore, Singapore 117411, Singapore
| |
Collapse
|
2
|
Zhao Q, Zhu Y, Ren Y, Yin S, Yu L, Huang R, Song S, Hu X, Zhu R, Cheng L, Xie N. Neurogenesis potential of oligodendrocyte precursor cells from oligospheres and injured spinal cord. Front Cell Neurosci 2022; 16:1049562. [PMID: 36619671 PMCID: PMC9813964 DOI: 10.3389/fncel.2022.1049562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Severe traumatic spinal cord injury (SCI) leads to long-lasting oligodendrocyte death and extensive demyelination in the lesion area. Oligodendrocyte progenitor cells (OPCs) are the reservoir of new mature oligodendrocytes during damaged myelin regeneration, which also have latent potential for neurogenic regeneration and oligospheres formation. Whether oligospheres derived OPCs can differentiate into neurons and the neurogenesis potential of OPCs after SCI remains unclear. In this study, primary OPCs cultures were used to generate oligospheres and detect the differentiation and neurogenesis potential of oligospheres. In vivo, SCI models of juvenile and adult mice were constructed. Combining the single-cell RNA sequencing (scRNA-seq), bulk RNA sequencing (RNA-seq), bioinformatics analysis, immunofluorescence staining, and molecular experiment, we investigated the neurogenesis potential and mechanisms of OPCs in vitro and vivo. We found that OPCs differentiation and oligodendrocyte morphology were significantly different between brain and spinal cord. Intriguingly, we identify a previously undescribed findings that OPCs were involved in oligospheres formation which could further differentiate into neuron-like cells. We also firstly detected the intermediate states of oligodendrocytes and neurons during oligospheres differentiation. Furthermore, we found that OPCs were significantly activated after SCI. Combining scRNA-seq and bulk RNA-seq data from injured spinal cord, we confirmed the neurogenesis potential of OPCs and the activation of endoplasmic reticulum stress after SCI. Inhibition of endoplasmic reticulum stress could effectively attenuate OPCs death. Additionally, we also found that endoplasmic reticulum may regulate the stemness and differentiation of oligospheres. These findings revealed the neurogenesis potential of OPCs from oligospheres and injured spinal cord, which may provide a new source and a potential target for spinal cord repair.
Collapse
Affiliation(s)
- Qing Zhao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China,Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Yanjing Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yilong Ren
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China,Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Shuai Yin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China,Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Liqun Yu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ruiqi Huang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Simin Song
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiao Hu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China,Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China,*Correspondence: Rongrong Zhu,
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China,Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China,Liming Cheng,
| | - Ning Xie
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China,Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China,Ning Xie,
| |
Collapse
|
3
|
Ji J, Yang C, Shan Y, Sun M, Cui X, Xu L, Liang S, Li T, Fan Y, Luo D, Li Z. Research Trends of Piezoelectric Nanomaterials in Biomedical Engineering. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Jianying Ji
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
- Center on Nanoenergy Research School of Physical Science and Technology Guangxi University Nanning 530004 China
| | - Chunyu Yang
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
- State Key Laboratory of Heavy Oil Processing College of New Energy and Materials Beijing Key Laboratory of Biogas Upgrading Utilization China University of Petroleum (Beijing) Beijing 102249 China
- Institute of Engineering Medicine School of Life Science Beijing Institute of Technology Beijing 100081 China
| | - Yizhu Shan
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
| | - Mingjun Sun
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
- State Key Laboratory of Heavy Oil Processing College of New Energy and Materials Beijing Key Laboratory of Biogas Upgrading Utilization China University of Petroleum (Beijing) Beijing 102249 China
- Institute of Engineering Medicine School of Life Science Beijing Institute of Technology Beijing 100081 China
| | - Xi Cui
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
| | - Lingling Xu
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
- National Center for Nanoscience and Technology Chinese Academy of Sciences Beijing 100190 China
| | - Shiyuan Liang
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
| | - Tong Li
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
- Center on Nanoenergy Research School of Physical Science and Technology Guangxi University Nanning 530004 China
| | - Yijie Fan
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
| | - Dan Luo
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
- School of Nanoscience and Technology University of Chinese Academy of Sciences Beijing 100049 China
| | - Zhou Li
- Institute of Nanoenergy and Nanosystems Chinese Academy of Science Beijing 101400 China
- Center on Nanoenergy Research School of Physical Science and Technology Guangxi University Nanning 530004 China
- School of Nanoscience and Technology University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
4
|
Yao L, Tran K, Nguyen D. Collagen Matrices Mediate Glioma Cell Migration Induced by an Electrical Signal. Gels 2022; 8:gels8090545. [PMID: 36135257 PMCID: PMC9498326 DOI: 10.3390/gels8090545] [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: 07/24/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Glioma cells produce an increased amount of collagen compared with normal astrocytes. The increasing amount of collagen in the extracellular matrix (ECM) modulates the matrix structure and the mechanical properties of the microenvironment, thereby regulating tumor cell invasion. Although the regulation of tumor cell invasion mainly relies on cell–ECM interaction, the electrotaxis of tumor cells has attracted great research interest. The growth of glioma cells in a three-dimensional (3D) collagen hydrogel creates a relevant tumor physiological condition for the study of tumor cell invasion. In this study, we tested the migration of human glioma cells, fetal astrocytes, and adult astrocytes in a 3D collagen matrix with different collagen concentrations. We report that all three types of cells demonstrated higher motility in a low concentration of collagen hydrogel (3 mg/mL and 5 mg/mL) than in a high concentration of collagen hydrogel (10 mg/mL). We further show that human glioma cells grown in collagen hydrogels responded to direct current electric field (dcEF) stimulation and migrated to the anodal pole. The tumor cells altered their morphology in the gels to adapt to the anodal migration. The directedness of anodal migration shows a field strength-dependent response. EF stimulation increased the migration speed of tumor cells. This study implicates the potential role of an dcEF in glioma invasion and as a target of treatment.
Collapse
Affiliation(s)
- Li Yao
- Correspondence: ; Tel.: +316-978-6766; Fax: +316-978-3772
| | | | | |
Collapse
|
5
|
Protocadherin 15 suppresses oligodendrocyte progenitor cell proliferation and promotes motility through distinct signalling pathways. Commun Biol 2022; 5:511. [PMID: 35637313 PMCID: PMC9151716 DOI: 10.1038/s42003-022-03470-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) express protocadherin 15 (Pcdh15), a member of the cadherin superfamily of transmembrane proteins. Little is known about the function of Pcdh15 in the central nervous system (CNS), however, Pcdh15 expression can predict glioma aggression and promote the separation of embryonic human OPCs immediately following a cell division. Herein, we show that Pcdh15 knockdown significantly increases extracellular signal-related kinase (ERK) phosphorylation and activation to enhance OPC proliferation in vitro. Furthermore, Pcdh15 knockdown elevates Cdc42-Arp2/3 signalling and impairs actin kinetics, reducing the frequency of lamellipodial extrusion and slowing filopodial withdrawal. Pcdh15 knockdown also reduces the number of processes supported by each OPC and new process generation. Our data indicate that Pcdh15 is a critical regulator of OPC proliferation and process motility, behaviours that characterise the function of these cells in the healthy CNS, and provide mechanistic insight into the role that Pcdh15 might play in glioma progression. Protocadherin 15 promotes lamellipodial and filopodial dynamics in oligodendrocyte progenitor cells by regulating Cdc42-Arp2/3 activity, but also suppresses ERK1/2 phosphorylation to reduce proliferation.
Collapse
|
6
|
Yoo DY, Jung HY, Kim W, Hahn KR, Kwon HJ, Nam SM, Chung JY, Yoon YS, Kim DW, Hwang IK. Entacapone Treatment Modulates Hippocampal Proteins Related to Synaptic Vehicle Trafficking. Cells 2020; 9:cells9122712. [PMID: 33352833 PMCID: PMC7765944 DOI: 10.3390/cells9122712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 11/29/2022] Open
Abstract
Entacapone, a reversible inhibitor of catechol-O-methyl transferase, is used for patients in Parkinson’s disease because it increases the bioavailability and effectiveness of levodopa. In the present study, we observed that entacapone increases novel object recognition and neuroblasts in the hippocampus. In the present study, two-dimensional electrophoresis (2-DE) and matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry were performed to compare the abundance profiles of proteins expressed in the hippocampus after entacapone treatment in mice. Results of 2-DE, MALDI-TOF mass spectrometry, and subsequent proteomic analysis revealed an altered protein expression profile in the hippocampus after entacapone treatment. Based on proteomic analysis, 556 spots were paired during the image analysis of 2-DE gels and 76 proteins were significantly changed more than two-fold among identified proteins. Proteomic analysis indicated that treatment with entacapone induced expressional changes in proteins involved in synaptic transmission, cellular processes, cellular signaling, the regulation of cytoskeletal structure, energy metabolism, and various subcellular enzymatic reactions. In particular, entacapone significantly increased proteins related to synaptic trafficking and plasticity, such as dynamin 1, synapsin I, and Munc18-1. Immunohistochemical staining showed the localization of the proteins, and western blot confirmed the significant increases in dynamin I (203.5% of control) in the hippocampus as well as synapsin I (254.0% of control) and Munc18-1 (167.1% of control) in the synaptic vesicle fraction of hippocampus after entacapone treatment. These results suggest that entacapone can enhance hippocampal synaptic trafficking and plasticity against various neurological diseases related to hippocampal dysfunction.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea;
| | - Sung Min Nam
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Iksan 54538, Korea;
| | - Jin Young Chung
- Department of Veterinary Internal Medicine and Geriatrics, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea;
- Correspondence: (D.W.K.); (I.K.H.)
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
- Correspondence: (D.W.K.); (I.K.H.)
| |
Collapse
|
7
|
Yao L, Shippy T, Li Y. Genetic analysis of the molecular regulation of electric fields-guided glia migration. Sci Rep 2020; 10:16821. [PMID: 33033380 PMCID: PMC7546725 DOI: 10.1038/s41598-020-74085-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/31/2020] [Indexed: 11/09/2022] Open
Abstract
In a developing nervous system, endogenous electric field (EF) influence embryonic growth. We reported the EF-directed migration of both rat Schwann cells (SCs) and oligodendrocyte precursor cells (OPCs) and explored the molecular mechanism using RNA-sequencing assay. However, previous studies revealed the differentially expressed genes (DEGs) associated with EF-guided migration of SCs or OPCs alone. In this study, we performed joint differential expression analysis on the RNA-sequencing data from both cell types. We report a number of significantly enriched gene ontology (GO) terms that are related to the cytoskeleton, cell adhesion, and cell migration. Of the DEGs associated with these terms, nine up-regulated DEGs and 32 down-regulated DEGs showed the same direction of effect in both SCs and OPCs stimulated with EFs, while the remaining DEGs responded differently. Thus, our study reveals the similarities and differences in gene expression and cell migration regulation of different glial cell types in response to EF stimulation.
Collapse
Affiliation(s)
- Li Yao
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA.
| | - Teresa Shippy
- Bioinformatics Specialist, KSU Bioinformatics Center, Kansas State University, Manhattan, KS, 66506, USA
| | - Yongchao Li
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA
| |
Collapse
|
8
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
9
|
Thomason EJ, Escalante M, Osterhout DJ, Fuss B. The oligodendrocyte growth cone and its actin cytoskeleton: A fundamental element for progenitor cell migration and CNS myelination. Glia 2019; 68:1329-1346. [PMID: 31696982 DOI: 10.1002/glia.23735] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/26/2019] [Accepted: 10/01/2019] [Indexed: 01/06/2023]
Abstract
Cells of the oligodendrocyte (OLG) lineage engage in highly motile behaviors that are crucial for effective central nervous system (CNS) myelination. These behaviors include the guided migration of OLG progenitor cells (OPCs), the surveying of local environments by cellular processes extending from differentiating and pre-myelinating OLGs, and during the process of active myelin wrapping, the forward movement of the leading edge of the myelin sheath's inner tongue along the axon. Almost all of these motile behaviors are driven by actin cytoskeletal dynamics initiated within a lamellipodial structure that is located at the tip of cellular OLG/OPC processes and is structurally as well as functionally similar to the neuronal growth cone. Accordingly, coordinated stoichiometries of actin filament (F-actin) assembly and disassembly at these OLG/OPC growth cones have been implicated in directing process outgrowth and guidance, and the initiation of myelination. Nonetheless, the functional importance of the OLG/OPC growth cone still remains to be fully understood, and, as a unique aspect of actin cytoskeletal dynamics, F-actin depolymerization and disassembly start to predominate at the transition from myelination initiation to myelin wrapping. This review provides an overview of the current knowledge about OLG/OPC growth cones, and it proposes a model in which actin cytoskeletal dynamics in OLG/OPC growth cones are a main driver for morphological transformations and motile behaviors. Remarkably, these activities, at least at the later stages of OLG maturation, may be regulated independently from the transcriptional gene expression changes typically associated with CNS myelination.
Collapse
Affiliation(s)
- Elizabeth J Thomason
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Miguel Escalante
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia.,Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Donna J Osterhout
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
10
|
Temporal and partial inhibition of GLI1 in neural stem cells (NSCs) results in the early maturation of NSC derived oligodendrocytes in vitro. Stem Cell Res Ther 2019; 10:272. [PMID: 31455382 PMCID: PMC6712625 DOI: 10.1186/s13287-019-1374-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 11/30/2022] Open
Abstract
Background Oligodendrocytes are a type of glial cells that synthesize the myelin sheath around the axons and are critical for the nerve conduction in the CNS. Oligodendrocyte death and defects are the leading causes of several myelin disorders such as multiple sclerosis, progressive multifocal leukoencephalopathy, periventricular leukomalacia, and several leukodystrophies. Temporal activation of the Sonic Hedgehog (SHH) pathway is critical for the generation of oligodendrocyte progenitors, and their differentiation and maturation in the brain and spinal cord during embryonic development in mammals. Methods Our protocol utilized adherent cultures of human induced pluripotent stem cells (iPSC) and human embryonic stem cells (hESCs) with a green fluorescent protein (GFP) reporter knocked into one allele of the OLIG2 gene locus, dual SMAD inhibition, and transient partial inhibition of glioma-associated oncogene 1 (GLI1) by the small molecule GANT61 during the formation of the SOX2/PAX6-positive neural stem cells (NSCs). The SHH pathway was later restimulated by a Smoothened agonist purmorphamine to induce the generation of OLIG2 glial precursors. One hundred ninety-two individual oligodendrocyte precursor cells (OPCs) from GANT61 and control group were analyzed by single-cell RNA sequencing (RNA-Seq). Results We demonstrate here that transient and partial inhibition of the SHH pathway transcription factor GLI1 in NSCs by a small molecule inhibitor GANT61 was found to generate OPCs that were more migratory and could differentiate earlier toward myelin-producing oligodendrocytes. Single-cell transcriptomic analysis (RNA-Seq) showed that GANT61-NSC-derived oligodendrocyte precursor cells (OPCs) had differential activation of some of the genes in the cytoskeleton rearrangement pathways that are involved in OPC motility and induction of maturation. At the protein level, this was also associated with higher levels of myelin-specific genes in the GANT61 group compared to controls. GANT61-NSC-derived OPCs were functional and could generate compact myelin in vitro and in vivo after transplantation in myelin-deficient shiverer mice. Conclusions This is a small molecule-based in vitro protocol that leads to the faster generation of functional oligodendrocytes. The development of protocols that lead to efficient and faster differentiation of oligodendrocytes from progenitors provides important advances toward the development of autologous neural stem cell-based therapies using human iPSCs. Electronic supplementary material The online version of this article (10.1186/s13287-019-1374-y) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Chen CY, Chou FS, Wang PS. Live-cell Migration Assays to Study Motility of Neural andGlial (Oligodendrocyte) Progenitor Cells. Bio Protoc 2019; 9:e3275. [PMID: 33654792 DOI: 10.21769/bioprotoc.3275] [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: 12/12/2018] [Revised: 05/22/2019] [Accepted: 06/26/2019] [Indexed: 11/02/2022] Open
Abstract
Cell motility has been extensively studied in in vitro models using fibroblasts and keratocytes, but the cell type-specific mechanisms underlying migration of lineage- or disease-specific cells, such as neural and glial progenitor cells, remain an active field for investigation. The migrating neural and glial progenitor cells contribute to the development, tissue repair and tumor invasion in the central nervous system (CNS). Cell migration is a highly dynamic process which relies on membranous protrusions to assemble, extend, disassemble and retract. In the CNS, the motility of neural and glial progenitor cells is affected by various cell-autonomous and non-cell-autonomous mechanisms such as signaling molecules, actin and microtubule interactions, and environmental cues. Here, we described a live-cell migration assay for use in the assessment of neural and glial progenitor cell migration. We first will demonstrate the procedures for isolating and culturing neural and glial progenitor cells. Next, we will demonstrate the acquisition of time-lapse images using phase contrast microscopy, the methods for quantification and the analyses of various motility parameters including speed, velocity, straightness and leading-edge dynamics. This method allows researchers to dissect the mechanisms of cell motility in response to different environmental cues, such as chemoattractive and repulsive signals, matrix adhesiveness and stiffness. This assay also allows researchers to study migration of pharmacologically and genetically manipulated cells.
Collapse
Affiliation(s)
- Chu-Yen Chen
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Fu-Sheng Chou
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA.,Division of Neonatology, Children's Mercy-Kansas City, Kansas City, MO, USA
| | - Pei-Shan Wang
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA
| |
Collapse
|
12
|
Dong ZY, Pei Z, Wang YL, Li Z, Khan A, Meng XT. Ascl1 Regulates Electric Field-Induced Neuronal Differentiation Through PI3K/Akt Pathway. Neuroscience 2019; 404:141-152. [PMID: 30771509 DOI: 10.1016/j.neuroscience.2019.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/14/2022]
Abstract
Directing differentiation of neural stem/progenitor cells (NSCs/NPCs) to produce functional neurons is one of the greatest challenges in regenerative medicine. Our previous paper has confirmed that electrical stimulation has a high efficiency of triggering neuronal differentiation by using isolated filum terminale (FT)-derived NPCs. To further clarify the intrinsic molecular mechanisms, protein-protein interaction (PPI) network analysis was applied to pinpoints novel hubs in electric field (EF)-induced neuronal differentiation. In this study, siRNA transfection of Achaete-scute homolog 1 (Ascl1) in NPCs or NPCs was followed by direct current stimulation at 150 mV/mm. Neuronal differentiation rate and protein expression level were analyzed after 7 or 14 days of electrical stimulation. The data showed that the expression level of Ascl1 was enhanced by electrical stimulation and positively correlated to EF strength. Moreover, we identified that the expression of Ascl1 positively regulated neuronal differentiation of NPCs and can be up-regulated by EF-stimulation through the activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway. Therefore, this study provides new insights into the role of Ascl1 and its relevant PI3K/Akt pathway in regulating of EF-induced neuronal differentiation and pointed out that continuous expression of Ascl1 in NPCs is required for EF-induced neuronal differentiation.
Collapse
Affiliation(s)
- Zhi-Yong Dong
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Zhe Pei
- Department of Neuroscience and Pediatric, GSRB1 Duke University, Durham 27710, USA
| | - Yan-Ling Wang
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Zhe Li
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Amber Khan
- The Graduate Center and CUNY School of Medicine, CUNY, 85 St Nicholas Terrace, New York, NY 10027, USA.
| | - Xiao-Ting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
13
|
Li J. Weak direct current (DC) electric fields as a therapy for spinal cord injuries: review and advancement of the oscillating field stimulator (OFS). Neurosurg Rev 2019; 42:825-834. [DOI: 10.1007/s10143-018-01068-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/07/2018] [Accepted: 12/07/2018] [Indexed: 12/14/2022]
|
14
|
Carvalho CR, Wrobel S, Meyer C, Brandenberger C, Cengiz IF, López-Cebral R, Silva-Correia J, Ronchi G, Reis RL, Grothe C, Oliveira JM, Haastert-Talini K. Gellan Gum-based luminal fillers for peripheral nerve regeneration: an in vivo study in the rat sciatic nerve repair model. Biomater Sci 2018; 6:1059-1075. [PMID: 29464240 DOI: 10.1039/c7bm01101f] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Peripheral nerve injuries (PNI) resulting in a gap to be bridged between the transected nerve ends are commonly reconstructed with autologous nerve tissue, but there is a need for valuable alternatives. This experimental work considers the innovative use of the biomaterial Gellan Gum (GG) as a luminal filler for nerve guidance channels made from chitosan with a 5% degree of acetylation. The engineered constructs should remodel the structural support given to regenerating axons by the so-called bands of Büngner. Four different GG formulations were produced by combining varying amounts of High-Acyl GG (HA-GG) and Methacrylated GG (MA-GG). The effective porosity of the freeze-dried networks was analysed by SEM and micro-CT 3D reconstructions, while the degradation and swelling abilities were characterized in vitro for up to 30 days. The metabolic activity and viability of immortalized Schwann cells seeded onto the freeze-dried networks were also evaluated. Finally, the developed hydrogel formulations were freeze-dried within the chitosan nerve guides and implanted in a 10 mm rat sciatic nerve defect. Functional and histomorphological analyses after 3, 6, and 12 weeks in vivo revealed that although it did not result in improved nerve regeneration, the NGC25:75 formulations could provide a basis for further development of GG scaffolds as luminal fillers for hollow nerve guidance channels.
Collapse
Affiliation(s)
- C R Carvalho
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque de Ciência e Tecnologia, Zona Industrial de Gandra, 4805-017 Barco, Guimarães, Portugal.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Yao L, Li Y. The Role of Direct Current Electric Field-Guided Stem Cell Migration in Neural Regeneration. Stem Cell Rev Rep 2017; 12:365-75. [PMID: 27108005 DOI: 10.1007/s12015-016-9654-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Effective directional axonal growth and neural cell migration are crucial in the neural regeneration of the central nervous system (CNS). Endogenous currents have been detected in many developing nervous systems. Experiments have demonstrated that applied direct current (DC) electric fields (EFs) can guide axonal growth in vitro, and attempts have been made to enhance the regrowth of damaged spinal cord axons using DC EFs in in vivo experiments. Recent work has revealed that the migration of stem cells and stem cell-derived neural cells can be guided by DC EFs. These studies have raised the possibility that endogenous and applied DC EFs can be used to direct neural tissue regeneration. Although the mechanism of EF-directed axonal growth and cell migration has not been fully understood, studies have shown that the polarization of cell membrane proteins and the activation of intracellular signaling molecules are involved in the process. The application of EFs is a promising biotechnology for regeneration of the CNS.
Collapse
Affiliation(s)
- Li Yao
- Department of Biological Sciences, Wichita State University, Wichita, KS, 67260, USA.
| | - Yongchao Li
- Department of Biological Sciences, Wichita State University, Wichita, KS, 67260, USA
| |
Collapse
|
16
|
Unraveling the mechanistic effects of electric field stimulation towards directing stem cell fate and function: A tissue engineering perspective. Biomaterials 2017; 150:60-86. [PMID: 29032331 DOI: 10.1016/j.biomaterials.2017.10.003] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/27/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
Electric field (EF) stimulation can play a vital role in eliciting appropriate stem cell response. Such an approach is recently being established to guide stem cell differentiation through osteogenesis/neurogenesis/cardiomyogenesis. Despite significant recent efforts, the biophysical mechanisms by which stem cells sense, interpret and transform electrical cues into biochemical and biological signals still remain unclear. The present review critically analyses the variety of EF stimulation approaches that can be employed to evoke appropriate stem cell response and also makes an attempt to summarize the underlying concepts of this notion, placing special emphasis on stem cell based tissue engineering and regenerative medicine. This review also discusses the major signaling pathways and cellular responses that are elicited by electric stimulation, including the participation of reactive oxygen species and heat shock proteins, modulation of intracellular calcium ion concentration, ATP production and numerous other events involving the clustering or reassembling of cell surface receptors, cytoskeletal remodeling and so on. The specific advantages of using external electric stimulation in different modalities to regulate stem cell fate processes are highlighted with explicit examples, in vitro and in vivo.
Collapse
|
17
|
Antal A, Alekseichuk I, Bikson M, Brockmöller J, Brunoni AR, Chen R, Cohen LG, Dowthwaite G, Ellrich J, Flöel A, Fregni F, George MS, Hamilton R, Haueisen J, Herrmann CS, Hummel FC, Lefaucheur JP, Liebetanz D, Loo CK, McCaig CD, Miniussi C, Miranda PC, Moliadze V, Nitsche MA, Nowak R, Padberg F, Pascual-Leone A, Poppendieck W, Priori A, Rossi S, Rossini PM, Rothwell J, Rueger MA, Ruffini G, Schellhorn K, Siebner HR, Ugawa Y, Wexler A, Ziemann U, Hallett M, Paulus W. Low intensity transcranial electric stimulation: Safety, ethical, legal regulatory and application guidelines. Clin Neurophysiol 2017; 128:1774-1809. [PMID: 28709880 PMCID: PMC5985830 DOI: 10.1016/j.clinph.2017.06.001] [Citation(s) in RCA: 718] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/29/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Abstract
Low intensity transcranial electrical stimulation (TES) in humans, encompassing transcranial direct current (tDCS), transcutaneous spinal Direct Current Stimulation (tsDCS), transcranial alternating current (tACS), and transcranial random noise (tRNS) stimulation or their combinations, appears to be safe. No serious adverse events (SAEs) have been reported so far in over 18,000 sessions administered to healthy subjects, neurological and psychiatric patients, as summarized here. Moderate adverse events (AEs), as defined by the necessity to intervene, are rare, and include skin burns with tDCS due to suboptimal electrode-skin contact. Very rarely mania or hypomania was induced in patients with depression (11 documented cases), yet a causal relationship is difficult to prove because of the low incidence rate and limited numbers of subjects in controlled trials. Mild AEs (MAEs) include headache and fatigue following stimulation as well as prickling and burning sensations occurring during tDCS at peak-to-baseline intensities of 1-2mA and during tACS at higher peak-to-peak intensities above 2mA. The prevalence of published AEs is different in studies specifically assessing AEs vs. those not assessing them, being higher in the former. AEs are frequently reported by individuals receiving placebo stimulation. The profile of AEs in terms of frequency, magnitude and type is comparable in healthy and clinical populations, and this is also the case for more vulnerable populations, such as children, elderly persons, or pregnant women. Combined interventions (e.g., co-application of drugs, electrophysiological measurements, neuroimaging) were not associated with further safety issues. Safety is established for low-intensity 'conventional' TES defined as <4mA, up to 60min duration per day. Animal studies and modeling evidence indicate that brain injury could occur at predicted current densities in the brain of 6.3-13A/m2 that are over an order of magnitude above those produced by tDCS in humans. Using AC stimulation fewer AEs were reported compared to DC. In specific paradigms with amplitudes of up to 10mA, frequencies in the kHz range appear to be safe. In this paper we provide structured interviews and recommend their use in future controlled studies, in particular when trying to extend the parameters applied. We also discuss recent regulatory issues, reporting practices and ethical issues. These recommendations achieved consensus in a meeting, which took place in Göttingen, Germany, on September 6-7, 2016 and were refined thereafter by email correspondence.
Collapse
Affiliation(s)
- A Antal
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany.
| | - I Alekseichuk
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - M Bikson
- Department of Biomedical Engineering, The City College of New York, New York, USA
| | - J Brockmöller
- Department of Clinical Pharmacology, University Medical Center Goettingen, Germany
| | - A R Brunoni
- Service of Interdisciplinary Neuromodulation, Department and Institute of Psychiatry, Laboratory of Neurosciences (LIM-27) and Interdisciplinary Center for Applied Neuromodulation University Hospital, University of São Paulo, São Paulo, Brazil
| | - R Chen
- Division of Neurology, Department of Medicine, University of Toronto and Krembil Research Institute, Toronto, Ontario, Canada
| | - L G Cohen
- Human Cortical Physiology and Neurorehabilitation Section, National Institute of Neurological Disorders and Stroke NIH, Bethesda, USA
| | | | - J Ellrich
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark; Institute of Physiology and Pathophysiology, University of Erlangen-Nürnberg, Erlangen, Germany; EBS Technologies GmbH, Europarc Dreilinden, Germany
| | - A Flöel
- Universitätsmedizin Greifswald, Klinik und Poliklinik für Neurologie, Greifswald, Germany
| | - F Fregni
- Spaulding Neuromodulation Center, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - M S George
- Brain Stimulation Division, Medical University of South Carolina, and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA
| | - R Hamilton
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - J Haueisen
- Institute of Biomedical Engineering and Informatics, Technische Universität Ilmenau, Germany
| | - C S Herrmann
- Experimental Psychology Lab, Department of Psychology, European Medical School, Carl von Ossietzky Universität, Oldenburg, Germany
| | - F C Hummel
- Defitech Chair of Clinical Neuroengineering, Centre of Neuroprosthetics (CNP) and Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland; Defitech Chair of Clinical Neuroengineering, Clinique Romande de Réadaptation, Swiss Federal Institute of Technology (EPFL Valais), Sion, Switzerland
| | - J P Lefaucheur
- Department of Physiology, Henri Mondor Hospital, Assistance Publique - Hôpitaux de Paris, and EA 4391, Nerve Excitability and Therapeutic Team (ENT), Faculty of Medicine, Paris Est Créteil University, Créteil, France
| | - D Liebetanz
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - C K Loo
- School of Psychiatry & Black Dog Institute, University of New South Wales, Sydney, Australia
| | - C D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - C Miniussi
- Center for Mind/Brain Sciences CIMeC, University of Trento, Rovereto, Italy; Cognitive Neuroscience Section, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - P C Miranda
- Institute of Biophysics and Biomedical Engineering, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - V Moliadze
- Institute of Medical Psychology and Medical Sociology, University Hospital of Schleswig-Holstein (UKSH), Campus Kiel, Christian-Albrechts-University, Kiel, Germany
| | - M A Nitsche
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany; Department of Neurology, University Hospital Bergmannsheil, Bochum, Germany
| | - R Nowak
- Neuroelectrics, Barcelona, Spain
| | - F Padberg
- Department of Psychiatry and Psychotherapy, Munich Center for Brain Stimulation, Ludwig-Maximilian University Munich, Germany
| | - A Pascual-Leone
- Division of Cognitive Neurology, Harvard Medical Center and Berenson-Allen Center for Noninvasive Brain Stimulation at Beth Israel Deaconess Medical Center, Boston, USA
| | - W Poppendieck
- Department of Information Technology, Mannheim University of Applied Sciences, Mannheim, Germany
| | - A Priori
- Center for Neurotechnology and Experimental Brain Therapeutich, Department of Health Sciences, University of Milan Italy; Deparment of Clinical Neurology, University Hospital Asst Santi Paolo E Carlo, Milan, Italy
| | - S Rossi
- Department of Medicine, Surgery and Neuroscience, Human Physiology Section and Neurology and Clinical Neurophysiology Section, Brain Investigation & Neuromodulation Lab, University of Siena, Italy
| | - P M Rossini
- Area of Neuroscience, Institute of Neurology, University Clinic A. Gemelli, Catholic University, Rome, Italy
| | | | - M A Rueger
- Department of Neurology, University Hospital of Cologne, Germany
| | | | | | - H R Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Y Ugawa
- Department of Neurology, Fukushima Medical University, Fukushima, Japan; Fukushima Global Medical Science Center, Advanced Clinical Research Center, Fukushima Medical University, Japan
| | - A Wexler
- Department of Science, Technology & Society, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - U Ziemann
- Department of Neurology & Stroke, and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - M Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - W Paulus
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| |
Collapse
|
18
|
Subcellular electrical stimulation of neurons enhances the myelination of axons by oligodendrocytes. PLoS One 2017; 12:e0179642. [PMID: 28671962 PMCID: PMC5495216 DOI: 10.1371/journal.pone.0179642] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 06/01/2017] [Indexed: 11/30/2022] Open
Abstract
Myelin formation has been identified as a modulator of neural plasticity. New tools are required to investigate the mechanisms by which environmental inputs and neural activity regulate myelination patterns. In this study, we demonstrate a microfluidic compartmentalized culture system with integrated electrical stimulation capabilities that can induce neural activity by whole cell and focal stimulation. A set of electric field simulations was performed to confirm spatial restriction of the electrical input in the compartmentalized culture system. We further demonstrate that electrode localization is a key consideration for generating uniform the stimulation of neuron and oligodendrocytes within the compartments. Using three configurations of the electrodes we tested the effects of subcellular activation of neural activity on distal axon myelination with oligodendrocytes. We further investigated if oligodendrocytes have to be exposed to the electrical field to induce axon myelination. An isolated stimulation of cell bodies and proximal axons had the same effect as an isolated stimulation of distal axons co-cultured with oligodendrocytes, and the two modes had a non-different result than whole cell stimulation. Our platform enabled the demonstration that electrical stimulation enhances oligodendrocyte maturation and myelin formation independent of the input localization and oligodendrocyte exposure to the electrical field.
Collapse
|
19
|
Samaddar S, Vazquez K, Ponkia D, Toruno P, Sahbani K, Begum S, Abouelela A, Mekhael W, Ahmed Z. Transspinal direct current stimulation modulates migration and proliferation of adult newly born spinal cells in mice. J Appl Physiol (1985) 2017; 122:339-353. [DOI: 10.1152/japplphysiol.00834.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/02/2016] [Accepted: 12/04/2016] [Indexed: 11/22/2022] Open
Abstract
Direct current electrical fields have been shown to be a major factor in the regulation of cell proliferation, differentiation, migration, and survival, as well as in the maturation of dividing cells during development. During adulthood, spinal cord cells are continuously produced in both animals and humans, and they hold great potential for neural restoration following spinal cord injury. While the effects of direct current electrical fields on adult-born spinal cells cultured ex vivo have recently been reported, the effects of direct current electrical fields on adult-born spinal cells in vivo have not been characterized. Here, we provide convincing findings that a therapeutic form of transspinal direct current stimulation (tsDCS) affects the migration and proliferation of adult-born spinal cells in mice. Specifically, cathodal tsDCS attracted the adult-born spinal cells, while anodal tsDCS repulsed them. In addition, both tsDCS polarities caused a significant increase in cell number. Regarding the potential mechanisms involved, both cathodal and anodal tsDCS caused significant increases in expression of brain-derived neurotrophic factor, while expression of nerve growth factor increased and decreased, respectively. In the spinal cord, both anodal and cathodal tsDCS increased blood flow. Since blood flow and angiogenesis are associated with the proliferation of neural stem cells, increased blood flow may represent a major factor in the modulation of newly born spinal cells by tsDCS. Consequently, we propose that the method and novel findings presented in the current study have the potential to facilitate cellular, molecular, and/or bioengineering strategies to repair injured spinal cords. NEW & NOTEWORTHY Our results indicate that transspinal direct current stimulation (tsDCS) affects the migratory pattern and proliferation of adult newly born spinal cells, a cell population which has been implicated in learning and memory. In addition, our results suggest a potential mechanism of action regarding the functional effects of applying direct current. Thus tsDCS may represent a novel method by which to manipulate the migration and cell number of adult newly born cells and restore functions following brain or spinal cord injury.
Collapse
Affiliation(s)
- Sreyashi Samaddar
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
| | - Kizzy Vazquez
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
| | - Dipen Ponkia
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
| | - Pedro Toruno
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
| | - Karim Sahbani
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
| | - Sultana Begum
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
| | - Ahmed Abouelela
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
| | - Wagdy Mekhael
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
- The Graduate Center, The City University of New York, New York, New York
| | - Zaghloul Ahmed
- Department of Physical Therapy, College of Staten Island Center for Developmental Neuroscience, Staten Island, New York; and
- The Graduate Center, The City University of New York, New York, New York
| |
Collapse
|
20
|
Zhu B, Nicholls M, Gu Y, Zhang G, Zhao C, Franklin RJM, Song B. Electric Signals Regulate the Directional Migration of Oligodendrocyte Progenitor Cells (OPCs) via β1 Integrin. Int J Mol Sci 2016; 17:ijms17111948. [PMID: 27879672 PMCID: PMC5133942 DOI: 10.3390/ijms17111948] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/29/2016] [Accepted: 11/11/2016] [Indexed: 01/13/2023] Open
Abstract
The guided migration of neural cells is essential for repair in the central nervous system (CNS). Oligodendrocyte progenitor cells (OPCs) will normally migrate towards an injury site to re-sheath demyelinated axons; however the mechanisms underlying this process are not well understood. Endogenous electric fields (EFs) are known to influence cell migration in vivo, and have been utilised in this study to direct the migration of OPCs isolated from neonatal Sprague-Dawley rats. The OPCs were exposed to physiological levels of electrical stimulation, and displayed a marked electrotactic response that was dependent on β1 integrin, one of the key subunits of integrin receptors. We also observed that F-actin, an important component of the cytoskeleton, was re-distributed towards the leading edge of the migrating cells, and that this asymmetric rearrangement was associated with β1 integrin function.
Collapse
Affiliation(s)
- Bangfu Zhu
- Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK.
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK.
| | - Matthew Nicholls
- Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK.
| | - Yu Gu
- Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK.
| | - Gaofeng Zhang
- Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK.
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 1TA, UK.
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 1TA, UK.
| | - Bing Song
- Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK.
- Department of Dermatology, No. 1 Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
21
|
Shortrede JE, Uzair ID, Neira FJ, Flamini MI, Sanchez AM. Paxillin, a novel controller in the signaling of estrogen to FAK/N-WASP/Arp2/3 complex in breast cancer cells. Mol Cell Endocrinol 2016; 430:56-67. [PMID: 27095481 DOI: 10.1016/j.mce.2016.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 11/25/2022]
Abstract
Breast cancer is the major cause of cancer-related death in women. Its treatment is particularly difficult when metastasis occurs. The ability of cancer cells to move and invade the surrounding environment is the basis of local and distant metastasis. Cancer cells are able to remodel the actin cytoskeleton, which requires the recruitment of numerous structural and regulatory proteins that modulate actin filaments dynamics, including Paxillin or the Neural Wiskott-Aldrich Syndrome Protein (N-WASP). We show that 17-β estradiol (E2) induces phosphorylation of Paxillin and its translocation toward membrane sites where focal adhesion complexes are assembled. This cascade is triggered by a Gαi1/Gβ protein-dependent signaling of estrogen receptor α (ERα) to c-Src, focal adhesion kinase (FAK) and Paxillin. Within this complex, activated Paxillin recruits the small GTPase Cdc42, which triggers N-WASP phosphorylation. This results in the redistribution of Arp2/3 complexes at sites where membrane structures related to cell movement are formed. Recruitment of Paxillin, Cdc42 and N-WASP is necessary for cell adhesion, migration and invasion induced by E2 in breast cancer cells. In parallel, we investigated whether Raloxifene (RAL), a selective estrogen receptor modulator (SERMs), could inhibit or revert the effects of E2 in breast cancer cell movement. We found that, in the presence of E2, RAL acts as an ER antagonist and displays an inhibitory effect on estrogen-promoted cell adhesion and migration via FAK/Paxillin/N-WASP. Our findings identify an original mechanism through which estrogen regulates breast cancer cell motility and invasion via Paxillin. These results may have clinical relevance for the development of new therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Jorge Eduardo Shortrede
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina
| | - Ivonne Denise Uzair
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina
| | - Flavia Judith Neira
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina
| | - Marina Inés Flamini
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina
| | - Angel Matías Sanchez
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina.
| |
Collapse
|
22
|
Wang PS, Chou FS, Ramachandran S, Xia S, Chen HY, Guo F, Suraneni P, Maher BJ, Li R. Crucial roles of the Arp2/3 complex during mammalian corticogenesis. Development 2016; 143:2741-52. [PMID: 27385014 PMCID: PMC5004905 DOI: 10.1242/dev.130542] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 06/20/2016] [Indexed: 02/06/2023]
Abstract
The polarity and organization of radial glial cells (RGCs), which serve as both stem cells and scaffolds for neuronal migration, are crucial for cortical development. However, the cytoskeletal mechanisms that drive radial glial outgrowth and maintain RGC polarity remain poorly understood. Here, we show that the Arp2/3 complex – the unique actin nucleator that produces branched actin networks – plays essential roles in RGC polarity and morphogenesis. Disruption of the Arp2/3 complex in murine RGCs retards process outgrowth toward the basal surface and impairs apical polarity and adherens junctions. Whereas the former is correlated with an abnormal actin-based leading edge, the latter is consistent with blockage in membrane trafficking. These defects result in altered cell fate, disrupted cortical lamination and abnormal angiogenesis. In addition, we present evidence that the Arp2/3 complex is a cell-autonomous regulator of neuronal migration. Our data suggest that Arp2/3-mediated actin assembly might be particularly important for neuronal cell motility in a soft or poorly adhesive matrix environment. Summary: During mouse cortical development, the Arp2/3 actin branching complex regulates process formation and the maintenance of radial glial cell polarity, as well as affecting neuronal migration.
Collapse
Affiliation(s)
- Pei-Shan Wang
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Fu-Sheng Chou
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave., Boston, MA 02115, USA
| | - Sreekumar Ramachandran
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Sheng Xia
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Huei-Ying Chen
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Fengli Guo
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Praveen Suraneni
- Division of Hematology/Oncology, Robert Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Brady J Maher
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, 4940 Eastern Ave., Baltimore, MD 21224, USA Department of Neuroscience, Johns Hopkins School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA
| | - Rong Li
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles St., Baltimore, MD 21218, USA
| |
Collapse
|
23
|
Swaney KF, Li R. Function and regulation of the Arp2/3 complex during cell migration in diverse environments. Curr Opin Cell Biol 2016; 42:63-72. [PMID: 27164504 DOI: 10.1016/j.ceb.2016.04.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023]
Abstract
As the first de novo actin nucleator discovered, the Arp2/3 complex has been a central player in models of protrusive force production via the dynamic actin network. Here, we review recent studies on the functional role of the Arp2/3 complex in the migration of diverse cell types in different migratory environments. These findings have revealed an unexpected level of plasticity, both in how cells rely on the Arp2/3 complex for migration and other physiological functions and in the intricate modulation of the Arp2/3 complex by other actin regulators and upstream signaling cascades.
Collapse
Affiliation(s)
- Kristen F Swaney
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, 450 Rangos Building, Baltimore, MD 21205, USA; Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, 3400 North Charles Street, 100 Croft Hall, Baltimore, MD 21218, USA.
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, 450 Rangos Building, Baltimore, MD 21205, USA; Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, 3400 North Charles Street, 100 Croft Hall, Baltimore, MD 21218, USA
| |
Collapse
|
24
|
Wang W, Townes-Anderson E. LIM Kinase, a Newly Identified Regulator of Presynaptic Remodeling by Rod Photoreceptors After Injury. Invest Ophthalmol Vis Sci 2016; 56:7847-58. [PMID: 26658506 DOI: 10.1167/iovs.15-17278] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Rod photoreceptors retract their axon terminals and develop neuritic sprouts in response to retinal detachment and reattachment, respectively. This study examines the role of LIM kinase (LIMK), a component of RhoA and Rac pathways, in the presynaptic structural remodeling of rod photoreceptors. METHODS Phosphorylated LIMK (p-LIMK), the active form of LIMK, was examined in salamander retina with Western blot and confocal microscopy. Axon length within the first 7 hours and process growth after 3 days of culture were assessed in isolated rod photoreceptors treated with inhibitors of upstream regulators ROCK and p21-activated kinase (Pak) (Y27632 and IPA-3) and a direct LIMK inhibitor (BMS-5). Porcine retinal explants were also treated with BMS-5 and analyzed 24 hours after detachment. Because Ca2+ influx contributes to axonal retraction, L-type channels were blocked in some experiments with nicardipine. RESULTS Phosphorylated LIMK is present in rod terminals during retraction and in newly formed processes. Axonal retraction over 7 hours was significantly reduced by inhibition of LIMK or its regulators, ROCK and Pak. Process growth was reduced by LIMK or Pak inhibition especially at the basal (axon-bearing) region of the rod cells. Combining Ca2+ channel and LIMK inhibition had no additional effect on retraction but did further inhibit sprouting after 3 days. In detached porcine retina, LIMK inhibition reduced rod axonal retraction and improved retinal morphology. CONCLUSIONS Thus structural remodeling, in the form of either axonal retraction or neuritic growth, requires LIMK activity. LIM kinase inhibition may have therapeutic potential for reducing pathologic rod terminal plasticity after retinal injury.
Collapse
|
25
|
Li Y, Wang X, Yao L. Directional migration and transcriptional analysis of oligodendrocyte precursors subjected to stimulation of electrical signal. Am J Physiol Cell Physiol 2015; 309:C532-40. [PMID: 26269459 DOI: 10.1152/ajpcell.00175.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/05/2015] [Indexed: 12/28/2022]
Abstract
Loss of oligodendrocytes as the result of central nervous system disease causes demyelination that impairs axon function. Effective directional migration of endogenous or grafted oligodendrocyte precursor cells (OPCs) to a lesion is crucial in the neural remyelination process. In this study, the migration of OPCs in electric fields (EFs) was investigated. We found that OPCs migrated anodally in applied EFs, and the directedness and displacement of anodal migration increased significantly when the EF strength increased from 50 to 200 mV/mm. However, EFs did not significantly affect the cell migration speed. The transcriptome of OPCs subjected to EF stimulation (100 and 200 mV/mm) was analyzed using RNA sequencing (RNA-Seq), and results were verified by the reverse transcription quantitative polymerase chain reaction. A Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that the mitogen-activated protein kinase pathway that signals cell migration was significantly upregulated in cells treated with an EF of 200 mV/mm compared with control cells. Gene ontology enrichment analysis showed the downregulation of differentially expressed genes in chemotaxis. This study suggests that an applied EF is an effective cue to guiding OPC migration in neural regeneration and that transcriptional analysis contributes to the understanding of the mechanism of EF-guided cell migration.
Collapse
Affiliation(s)
- Yongchao Li
- Department of Biological Sciences, Wichita State University, Wichita, Kansas; and
| | - Xinkun Wang
- Genome Sequencing Core and Genomics Facility, University of Kansas, Lawrence, Kansas
| | - Li Yao
- Department of Biological Sciences, Wichita State University, Wichita, Kansas; and
| |
Collapse
|