1
|
Yamakawa M, Santosa SM, Chawla N, Ivakhnitskaia E, Del Pino M, Giakas S, Nadel A, Bontu S, Tambe A, Guo K, Han KY, Cortina MS, Yu C, Rosenblatt MI, Chang JH, Azar DT. Transgenic models for investigating the nervous system: Currently available neurofluorescent reporters and potential neuronal markers. Biochim Biophys Acta Gen Subj 2020; 1864:129595. [PMID: 32173376 DOI: 10.1016/j.bbagen.2020.129595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Recombinant DNA technologies have enabled the development of transgenic animal models for use in studying a myriad of diseases and biological states. By placing fluorescent reporters under the direct regulation of the promoter region of specific marker proteins, these models can localize and characterize very specific cell types. One important application of transgenic species is the study of the cytoarchitecture of the nervous system. Neurofluorescent reporters can be used to study the structural patterns of nerves in the central or peripheral nervous system in vivo, as well as phenomena involving embryologic or adult neurogenesis, injury, degeneration, and recovery. Furthermore, crucial molecular factors can also be screened via the transgenic approach, which may eventually play a major role in the development of therapeutic strategies against diseases like Alzheimer's or Parkinson's. This review describes currently available reporters and their uses in the literature as well as potential neural markers that can be leveraged to create additional, robust transgenic models for future studies.
Collapse
Affiliation(s)
- Michael Yamakawa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Samuel M Santosa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Neeraj Chawla
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Evguenia Ivakhnitskaia
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Matthew Del Pino
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Sebastian Giakas
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Arnold Nadel
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Sneha Bontu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Arjun Tambe
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Kai Guo
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Maria Soledad Cortina
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Charles Yu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America.
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America.
| |
Collapse
|
2
|
Smolders SMT, Kessels S, Vangansewinkel T, Rigo JM, Legendre P, Brône B. Microglia: Brain cells on the move. Prog Neurobiol 2019; 178:101612. [PMID: 30954517 DOI: 10.1016/j.pneurobio.2019.04.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/13/2019] [Accepted: 04/01/2019] [Indexed: 02/08/2023]
Abstract
In the last decade, tremendous progress has been made in understanding the biology of microglia - i.e. the fascinating immigrated resident immune cell population of the central nervous system (CNS). Recent literature reviews have largely dealt with the plentiful functions of microglia in CNS homeostasis, development and pathology, and the influences of sex and the microbiome. In this review, the intriguing aspect of their physical plasticity during CNS development will get specific attention. Microglia move around (mobility) and reshape their processes (motility). Microglial migration into and inside the CNS is most prominent throughout development and consequently most of the data described in this review concern mobility and motility in the changing environment of the developing brain. Here, we first define microglia based on their highly specialized age- and region-dependent gene expression signature and associated functional heterogeneity. Next, we describe their origin, the migration route of immature microglial cells towards the CNS, the mechanisms underlying their invasion of the CNS, and their spatiotemporal localization and surveying behaviour inside the developing CNS. These processes are dependent on microglial mobility and motility which are determined by the microenvironment of the CNS. Therefore, we further zoom in on the changing environment during CNS development. We elaborate on the extracellular matrix and the respective integrin receptors on microglia and we discuss the purinergic and molecular signalling in microglial mobility. In the last section, we discuss the physiological and pathological functions of microglia in which mobility and motility are involved to stress the importance of microglial 'movement'.
Collapse
Affiliation(s)
- Sophie Marie-Thérèse Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium; INSERM, UMR-S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France; Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | | | | | - Pascal Legendre
- INSERM, UMR-S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France; Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | |
Collapse
|
3
|
Smolders SMT, Swinnen N, Kessels S, Arnauts K, Smolders S, Le Bras B, Rigo JM, Legendre P, Brône B. Age-specific function of α5β1 integrin in microglial migration during early colonization of the developing mouse cortex. Glia 2017; 65:1072-1088. [PMID: 28417486 DOI: 10.1002/glia.23145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
Abstract
Microglia, the immune cells of the central nervous system, take part in brain development and homeostasis. They derive from primitive myeloid progenitors that originate in the yolk sac and colonize the brain mainly through intensive migration. During development, microglial migration speed declines which suggests that their interaction with the microenvironment changes. However, the matrix-cell interactions allowing dispersion within the parenchyma are unknown. Therefore, we aimed to better characterize the migration behavior and to assess the role of matrix-integrin interactions during microglial migration in the embryonic brain ex vivo. We focused on microglia-fibronectin interactions mediated through the fibronectin receptor α5β1 integrin because in vitro work indirectly suggested a role for this ligand-receptor pair. Using 2-photon time-lapse microscopy on acute ex vivo embryonic brain slices, we found that migration occurs in a saltatory pattern and is developmentally regulated. Most importantly, there is an age-specific function of the α5β1 integrin during microglial cortex colonization. At embryonic day (E) 13.5, α5β1 facilitates migration while from E15.5, it inhibits migration. These results indicate a developmentally regulated function of α5β1 integrin in microglial migration during colonization of the embryonic brain.
Collapse
Affiliation(s)
- Sophie Marie-Thérèse Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium.,INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | | | | | - Silke Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium.,Laboratory of Neuronal Differentiation, VIB Center for the Biology of Disease, Leuven and Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Barbara Le Bras
- INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | - Pascal Legendre
- INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | |
Collapse
|
4
|
Neuronal production from induced pluripotent stem cells in self-assembled collagen-hyaluronic acid-alginate microgel scaffolds with grafted GRGDSP/Ln5-P4. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:760-774. [PMID: 28482588 DOI: 10.1016/j.msec.2017.03.133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 01/22/2023]
Abstract
Self-assembled microgel functionalized with peptides was developed and applied to regenerate neurons from induced pluripotent stem cells (iPSCs). Collagen (COL), hyaluronic acid (HA), and alginate (ALG) were modified with methacrylic anhydride (MA), photocrosslinked for patterned particles, grafted with GRGDSP and Ln5-P4, and self-assembled to integrate the microgel into three-dimensional scaffolds. Physicochemical assessments revealed that the ternary microgel scaffolds had an optimal chemical composition at COLMA:HAMA:ALGMA=1:2:1. In fabricating cell-laden constructs, modified GRGDSP/Ln5-P4 in linear self-assembled scaffolds could significantly improve the entrapment efficiency and viability of iPSCs. In addition, GRGDSP/Ln5-P4 in the microgel constructs triggered the differentiation of iPSCs toward neurons, since the percentage of neurite-like cells could be higher than 98% after induction of nerve growth factor. Self-assembled microgel comprising COLMA, HAMA, ALGMA, and GRGDSP/Ln5-P4 may be promising in producing mature neural lineage from iPSCs, to provide better treatment for damaged nervous tissue.
Collapse
|
5
|
Zeng X, Ma YH, Chen YF, Qiu XC, Wu JL, Ling EA, Zeng YS. Autocrine fibronectin from differentiating mesenchymal stem cells induces the neurite elongation in vitro and promotes nerve fiber regeneration in transected spinal cord injury. J Biomed Mater Res A 2016; 104:1902-11. [PMID: 26991461 PMCID: PMC5101622 DOI: 10.1002/jbm.a.35720] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 03/02/2016] [Accepted: 03/11/2016] [Indexed: 01/08/2023]
Abstract
Extracellular matrix (ECM) expression is temporally and spatially regulated during the development of stem cells. We reported previously that fibronectin (FN) secreted by bone marrow mesenchymal stem cells (MSCs) was deposited on the surface of gelatin sponge (GS) soon after culture. In this study, we aimed to assess the function of accumulated FN on neuronal differentiating MSCs as induced by Schwann cells (SCs) in three dimensional transwell co‐culture system. The expression pattern and amount of FN of differentiating MSCs was examined by immunofluorescence, Western blot and immunoelectron microscopy. The results showed that FN accumulated inside GS scaffold, although its mRNA expression in MSCs was progressively decreased during neural induction. MSC‐derived neuron‐like cells showed spindle‐shaped cell body and long extending processes on FN‐decorated scaffold surface. However, after blocking of FN function by application of monoclonal antibodies, neuron‐like cells showed flattened cell body with short and thick neurites, together with decreased expression of integrin β1. In vivo transplantation study revealed that autocrine FN significantly facilitated endogenous nerve fiber regeneration in spinal cord transection model. Taken together, the present results showed that FN secreted by MSCs in the early stage accumulated on the GS scaffold and promoted the neurite elongation of neuronal differentiating MSCs as well as nerve fiber regeneration after spinal cord injury. This suggests that autocrine FN has a dynamic influence on MSCs in a three dimensional culture system and its potential application for treatment of traumatic spinal cord injury. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1902–1911, 2016.
Collapse
Affiliation(s)
- Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
- Department of Histology and Embryology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yuan-Feng Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xue-Cheng Qiu
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
| | - Jin-Lang Wu
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
- Co-innovation Center of Neuroregeneration, Jiangsu, 226019, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
6
|
Yao S, Liu X, He J, Wang X, Wang Y, Cui FZ. Ordered self-assembled monolayers terminated with different chemical functional groups direct neural stem cell linage behaviours. Biomed Mater 2015; 11:014107. [DOI: 10.1088/1748-6041/11/1/014107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
7
|
Skop NB, Calderon F, Cho CH, Gandhi CD, Levison SW. Optimizing a multifunctional microsphere scaffold to improve neural precursor cell transplantation for traumatic brain injury repair. J Tissue Eng Regen Med 2013; 10:E419-E432. [DOI: 10.1002/term.1832] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 06/12/2013] [Accepted: 09/02/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Nolan B. Skop
- Department of Neurology and Neurosciences; Rutgers-New Jersey Medical School; Newark NJ USA
- Department of Neurological Surgery; Rutgers-New Jersey Medical School; Newark NJ USA
| | - Frances Calderon
- Department of Neurology and Neurosciences; Rutgers-New Jersey Medical School; Newark NJ USA
| | - Cheul H. Cho
- Department of Biomedical Engineering; New Jersey Institute of Technology; Newark NJ USA
| | - Chirag D. Gandhi
- Department of Neurological Surgery; Rutgers-New Jersey Medical School; Newark NJ USA
| | - Steven W. Levison
- Department of Neurology and Neurosciences; Rutgers-New Jersey Medical School; Newark NJ USA
| |
Collapse
|
8
|
Rychly J. Biointerface Technology. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
9
|
Amemiya Y, Kawano K, Matsusaki M, Akashi M, Nakamura N, Nakamura C. Formation of nanofilms on cell surfaces to improve the insertion efficiency of a nanoneedle into cells. Biochem Biophys Res Commun 2012; 420:662-5. [PMID: 22450311 DOI: 10.1016/j.bbrc.2012.03.062] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/12/2012] [Indexed: 11/28/2022]
Abstract
A nanoneedle, an atomic force microscope (AFM) tip etched to 200 nm in diameter and 10 μm in length, can be inserted into cells with the aid of an AFM and has been used to introduce functional molecules into cells and to analyze intracellular information with minimal cell damage. However, some cell lines have shown low insertion efficiency of the nanoneedle. Improvement in the insertion efficiency of a nanoneedle into such cells is a significant issue for nanoneedle-based cell manipulation and analysis. Here, we have formed nanofilms composed of extracellular matrix molecules on cell surfaces and found that the formation of the nanofilms improved insertion efficiency of a nanoneedle into fibroblast and neural cells. The nanofilms were shown to improve insertion efficiency even in cells in which the formation of actin stress fibers was inhibited by the ROCK inhibitor Y27632, suggesting that the nanofilms with the mesh structure directly contributed to the improved insertion efficiency of a nanoneedle.
Collapse
Affiliation(s)
- Yosuke Amemiya
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8562, Japan
| | | | | | | | | | | |
Collapse
|
10
|
Tonge DA, de Burgh HT, Docherty R, Humphries MJ, Craig SE, Pizzey J. Fibronectin supports neurite outgrowth and axonal regeneration of adult brain neurons in vitro. Brain Res 2012; 1453:8-16. [PMID: 22483961 PMCID: PMC3989037 DOI: 10.1016/j.brainres.2012.03.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 03/03/2012] [Accepted: 03/09/2012] [Indexed: 12/09/2022]
Abstract
The molecular basis of axonal regeneration of central nervous system (CNS) neurons remains to be fully elucidated. In part, this is due to the difficulty in maintaining CNS neurons in vitro. Here, we show that dissociated neurons from the cerebral cortex and hippocampus of adult mice may be maintained in culture for up to 9 days in defined medium without added growth factors. Outgrowth of neurites including axons was observed from both CNS sources and was significantly greater on plasma fibronectin than on other substrata such as laminin and merosin. Neurite outgrowth on fibronectin appears to be mediated by α5β1 integrin since a recombinant fibronectin fragment containing binding sites for this receptor was as effective as intact fibronectin in supporting neurite outgrowth. Conversely, function-blocking antibodies to α5 and β1 integrin sub-units inhibited neurite outgrowth on intact fibronectin. These results suggest that the axonal regeneration seen in in vivo studies using fibronectin-based matrices is due to the molecule itself and not a consequence of secondary events such as cellular infiltration. They also indicate the domains of fibronectin that may be responsible for eliciting this response.
Collapse
Affiliation(s)
- David A Tonge
- Wolfson-Centre for Age-Related Disease, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, UK
| | | | | | | | | | | |
Collapse
|
11
|
Myers JP, Santiago-Medina M, Gomez TM. Regulation of axonal outgrowth and pathfinding by integrin-ECM interactions. Dev Neurobiol 2011; 71:901-23. [PMID: 21714101 PMCID: PMC3192254 DOI: 10.1002/dneu.20931] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Developing neurons use a combination of guidance cues to assemble a functional neural network. A variety of proteins immobilized within the extracellular matrix (ECM) provide specific binding sites for integrin receptors on neurons. Integrin receptors on growth cones associate with a number of cytosolic adaptor and signaling proteins that regulate cytoskeletal dynamics and cell adhesion. Recent evidence suggests that soluble growth factors and classic axon guidance cues may direct axon pathfinding by controlling integrin-based adhesion. Moreover, because classic axon guidance cues themselves are immobilized within the ECM and integrins modulate cellular responses to many axon guidance cues, interactions between activated receptors modulate cell signals and adhesion. Ultimately, growth cones control axon outgrowth and pathfinding behaviors by integrating distinct biochemical signals to promote the proper assembly of the nervous system. In this review, we discuss our current understanding how ECM proteins and their associated integrin receptors control neural network formation.
Collapse
Affiliation(s)
- Jonathan P Myers
- Department of Neuroscience, Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
12
|
Rychly J. Biointerface Technology. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
13
|
Nishikawa K, Ayukawa K, Hara Y, Wada K, Aoki S. Endothelin/endothelin-B receptor signals regulate ventricle-directed interkinetic nuclear migration of cerebral cortical neural progenitors. Neurochem Int 2010; 58:261-72. [PMID: 21130129 DOI: 10.1016/j.neuint.2010.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 11/16/2010] [Accepted: 11/22/2010] [Indexed: 11/16/2022]
Abstract
We determined the expression profile of ∼300 G protein-coupled receptors (GPCRs) in embryonic cortical neural progenitor cells (NPCs) and identified a number of highly expressed GPCRs, among which endothelin-B receptor (ET(B)-R) was expressed at the highest level. We also revealed that endothelins (ETs) were predominantly expressed in CD31-positive endothelial cells of the embryonic cerebral cortex. Activation of ET(B)-R induced NPC assembly in vitro by promoting fibronectin-dependent-motility and N-cadherin-associated cell contact. NPC assembly also required a Rho-family GTPase(s) and phosphatidylinositol-3-kinase. In the embryonic cerebral cortex, a specific ET(B)-R agonist, IRL-1620, accelerated interkinetic nuclear migration (INM) of NPCs toward the ventricular wall (VW) ex vivo. Conversely, a specific ET(B)-R antagonist, BQ788, slowed INM, thereby inducing mislocalization of phospho-histone H3-positive M-phase nuclei in the ventricular zone (VZ) and decreasing the number of Tuj1-positive newborn neurons. Our results suggest that ET(B)-R-mediated assembly signals drive INM that precedes neurogenesis.
Collapse
Affiliation(s)
- Kaori Nishikawa
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8502, Japan
| | | | | | | | | |
Collapse
|
14
|
Fietz SA, Kelava I, Vogt J, Wilsch-Bräuninger M, Stenzel D, Fish JL, Corbeil D, Riehn A, Distler W, Nitsch R, Huttner WB. OSVZ progenitors of human and ferret neocortex are epithelial-like and expand by integrin signaling. Nat Neurosci 2010; 13:690-9. [PMID: 20436478 DOI: 10.1038/nn.2553] [Citation(s) in RCA: 585] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 04/19/2010] [Indexed: 12/18/2022]
Abstract
A major cause of the cerebral cortex expansion that occurred during evolution is the increase in subventricular zone (SVZ) progenitors. We found that progenitors in the outer SVZ (OSVZ) of developing human neocortex retain features of radial glia, in contrast to rodent SVZ progenitors, which have limited proliferation potential. Although delaminating from apical adherens junctions, OSVZ progenitors maintained a basal process contacting the basal lamina, a canonical epithelial property. OSVZ progenitor divisions resulted in asymmetric inheritance of their basal process. Notably, OSVZ progenitors are also found in the ferret, a gyrencephalic nonprimate. Functional disruption of integrins, expressed on the basal process of ferret OSVZ progenitors, markedly decreased the OSVZ progenitor population size, but not that of other, process-lacking SVZ progenitors, in slice cultures of ferret neocortex. Our findings suggest that maintenance of this epithelial property allows integrin-mediated, repeated asymmetric divisions of OSVZ progenitors, providing a basis for neocortical expansion.
Collapse
Affiliation(s)
- Simone A Fietz
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Physicochemical control of adult stem cell differentiation: shedding light on potential molecular mechanisms. J Biomed Biotechnol 2010; 2010:743476. [PMID: 20379388 PMCID: PMC2850549 DOI: 10.1155/2010/743476] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 01/27/2010] [Indexed: 12/15/2022] Open
Abstract
Realization of the exciting potential for stem-cell-based biomedical and therapeutic applications, including tissue engineering, requires an understanding of the cell-cell and cell-environment interactions. To this end, recent efforts have been focused on the manipulation of adult stem cell differentiation using inductive soluble factors, designing suitable mechanical environments, and applying noninvasive physical forces. Although each of these different approaches has been successfully applied to regulate stem cell differentiation, it would be of great interest and importance to integrate and optimally combine a few or all of the physicochemical differentiation cues to induce synergistic stem cell differentiation. Furthermore, elucidation of molecular mechanisms that mediate the effects of multiple differentiation cues will enable the researcher to better manipulate stem cell behavior and response.
Collapse
|
16
|
Meland MN, Herndon ME, Stipp CS. Expression of alpha5 integrin rescues fibronectin responsiveness in NT2N CNS neuronal cells. J Neurosci Res 2010; 88:222-32. [PMID: 19598247 DOI: 10.1002/jnr.22171] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The extracellular matrix protein fibronectin is implicated in neuronal regeneration in the peripheral nervous system. In the central nervous system (CNS), fibronectin is up-regulated at sites of penetrating injuries and stroke; however, CNS neurons down-regulate the fibronectin receptor alpha5beta1 integrin during differentiation and generally respond poorly to fibronectin. NT2N CNS neuron-like cells (derived from NT2 precursor cells) have been used in preclinical and clinical studies for treatment of stroke and a variety of CNS injury and disease models. Here we show that, like primary CNS neurons, NT2N cells down-regulate alpha5beta1 integrin during differentiation and respond poorly to fibronectin. The poor neurite outgrowth by NT2N cells on fibronectin can be rescued by transducing NT2 precursors with a retroviral vector expressing alpha5 integrin under the control of the murine stem cell virus 5' long terminal repeat. Sustained alpha5 integrin expression is compatible with the CNS-like neuronal differentiation of NT2N cells and does not prevent robust neurite outgrowth on other integrin ligands. Thus, alpha5 integrin expression in CNS neuronal precursor cells may provide a strategy for enhancing the outgrowth and survival of implanted cells in cell-replacement therapies for CNS injury and disease.
Collapse
Affiliation(s)
- Marit N Meland
- Department of Biology, University of Iowa, Iowa City, Iowa, USA
| | | | | |
Collapse
|
17
|
Marchetti G, Escuin S, van der Flier A, De Arcangelis A, Hynes RO, Georges-Labouesse E. Integrin alpha5beta1 is necessary for regulation of radial migration of cortical neurons during mouse brain development. Eur J Neurosci 2010; 31:399-409. [PMID: 20105241 PMCID: PMC3460545 DOI: 10.1111/j.1460-9568.2009.07072.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
During cerebral cortex development, post-mitotic neurons interact with radial glial fibers and the extracellular environment to migrate away from the ventricular region and form a correct laminar structure. Integrin receptors are major mediators of cell–cell and cell–extracellular matrix interactions. Several integrin heterodimers are present during formation of the cortical layers. The α5β1 receptor is expressed in the neural progenitors of the ventricular zone during cerebral cortex formation. Using in utero electroporation to introduce short hairpin RNAs in the brain at embryonic day 15.5, we were able to inhibit acutely the expression of α5 integrin in the developing cortex. The knockdown of α5 integrin expression level in neural precursors resulted in an inhibition of radial migration, without perturbing the glial scaffold. Moreover, the same inhibitory effect on neuronal migration was observed after electroporation of a Cre recombinase expression plasmid into the neural progenitors of conditional knockout mice for α5 integrin. In both types of experiments, the electroporated cells expressing reduced levels of α5 integrin accumulated in the premigratory region with an abnormal morphology. At postnatal day 2, ectopic neurons were observed in cortical layer V, while a deficit of neurons was observed in cortical layer II–IV. We show that these neurons do not express a layer V-specific marker, suggesting that they have not undergone premature differentiation. Overall, these results indicate that α5β1 integrin functions in the regulation of neural morphology and migration during cortical development, playing a role in cortical lamination.
Collapse
Affiliation(s)
- Giovanni Marchetti
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Cell Biology and Development, CNRS UMR7104, Inserm U964, Université de Strasbourg, 67404 Illkirch, France
| | | | | | | | | | | |
Collapse
|
18
|
Harper MM, Ye EA, Blong CC, Jacobson ML, Sakaguchi DS. Integrins contribute to initial morphological development and process outgrowth in rat adult hippocampal progenitor cells. J Mol Neurosci 2009; 40:269-83. [PMID: 19499350 DOI: 10.1007/s12031-009-9211-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Accepted: 05/19/2009] [Indexed: 10/20/2022]
Abstract
Adult rat hippocampal progenitor cells (AHPCs) are self-renewing, multipotent neural progenitor cells (NPCs) that can differentiate into neurons, oligodendrocytes, and astrocytes. AHPCs contact a variety of molecular cues within their surrounding microenvironment via integrins. We hypothesize that integrin receptors are important for NPCs. In this study, we have examined the distribution of integrins in neuronal-like, oligodendrocyte-like, and astrocyte-like AHPCs when grown on substrates that support integrin-mediated adhesion (laminin, fibronectin), and those that do not (poly-L: -ornithine, PLO) using immunocytochemistry as well as characterized the phenotypic differentiation of AHPCs plated on laminin and fibronectin. Focal adhesions were prominent in AHPCs plated on purified substrates, but were also found in AHPCs plated on PLO. The focal adhesions observed in AHPCs plated on PLO substrates may be formed by self-adhesion to the endogenously produced laminin or fibronectin. We have demonstrated that integrins contribute to the initial morphological differentiation of AHPCs, as inhibition of fibronectin binding with the competitive inhibitor echistatin significantly decreased the number of processes and microspikes present in treated cells, and also decreased overall cell area. Finally, we have characterized the genetic profile of a subset of integrins and integrin-related genes in the AHPCs using reverse transcriptase polymerase chain reaction. These results demonstrate an important role of integrins, in vitro, for the initial morphological differentiation of AHPCs.
Collapse
|
19
|
Tanaka Y, Tozuka Y, Takata T, Shimazu N, Matsumura N, Ohta A, Hisatsune T. Excitatory GABAergic activation of cortical dividing glial cells. ACTA ACUST UNITED AC 2009; 19:2181-95. [PMID: 19131437 DOI: 10.1093/cercor/bhn238] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Adult neocortex contains dividing satellite glia population even though their characteristics and functions have still remained unknown. Nestin(+)/NG2(+) cells as major fraction of dividing glial cells express bicuculline-sensitive gamma-aminobutyric acid A (GABA(A)) receptors and receive GABAergic inputs. Due to their high [Cl(-)](i), GABAergic activation depolarized the cells and then induced Ca(2+) influx into them. To assess an effect of this GABAergic excitation, we looked for the expression of neurotrophic factors. Among them, we detected the expression of brain-derived neurotrophic factor (BDNF) on the cells. The level of BDNF expression was elevated after cortical ischemia, and this elevation was blocked by bumetanide, an inhibitor for NKCC1 that blocks the GABAergic depolarization. Furthermore, performing a modified adhesive removal test, we observed that the treatment of bumetanide significantly attenuated the recovery in somatosensory dysfunction. Our results may shed a light on satellite glia population in the cortex and imply their roles in the functional recovery after ischemic injuries.
Collapse
Affiliation(s)
- Yuichi Tanaka
- Department of Integrated Biosciences, University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Park JH, Ahn JI, Kim SY, Park KS, Lee YD, Yamaguchi M, Chung HJ. Genetic modification does not affect the stemness of neural stem cells in nestin promoter-GFP transgenic mice. Neurosci Lett 2007; 421:185-90. [PMID: 17574339 DOI: 10.1016/j.neulet.2007.02.069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 01/31/2007] [Accepted: 02/19/2007] [Indexed: 10/23/2022]
Abstract
Because nestin promoter-GFP mice have frequently been used in neural stem cell (NSC) research, it is essential to prove that there is no alteration in the stemness of NSCs derived from this transgenic model for the interpretation and validity of the data. We compared the stemness of NSCs derived from transgenic mice expressing GFP driven by the nestin enhancer with those from wild-type (C57BL/6) mice with respect to the general gene expression profile, expression of neural stem cell markers as nestin and Sox2, and responsiveness to neurotrophins (BDNF, PDGF-BB, and NT-3). The gene expression profile analysis showed that the coefficient of correlation between the two groups was very high (r=0.9865) in the total genes. We found that 23 genes were either up- or down-regulated more than two-fold in the NSCs from the transgenic mice (p<0.05), without any obvious functional relatedness among them. Likewise, there was no difference between the two mouse groups in the expression of nestin or Sox2, the ability to form neurospheres and the neuronal differentiation of NSCs by neurotrophins. Taken together, the self-renewal and neuronal differentiation ability of NSCs from the transgenic mice showed the great similarity to those from wild-type mice. Such information will be useful when the properties of NSCs are evaluated following genetic modification in such a nestin-GFP Tg model.
Collapse
Affiliation(s)
- Jae Hyun Park
- Department of Pharmacology, National Institute of Toxicological Research, 5 Nokbun-Dong, Eunpyung-Gu, Seoul 122-704, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
21
|
Chen H, Titushkin I, Stroscio M, Cho M. Altered membrane dynamics of quantum dot-conjugated integrins during osteogenic differentiation of human bone marrow derived progenitor cells. Biophys J 2006; 92:1399-408. [PMID: 17114225 PMCID: PMC1783865 DOI: 10.1529/biophysj.106.094896] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Functionalized quantum dots offer several advantages for tracking the motion of individual molecules on the cell surface, including selective binding, precise optical identification of cell surface molecules, and detailed examination of the molecular motion without photobleaching. We have used quantum dots conjugated with integrin antibodies and performed studies to quantitatively demonstrate changes in the integrin dynamics during osteogenic differentiation of human bone marrow derived progenitor cells (BMPCs). Consistent with the unusually strong BMPC adhesion previously observed, integrins on the surface of undifferentiated BMPC were found in clusters and the lateral diffusion was slow (e.g., approximately 10(-11) cm2/s). At times as early as those after a 3-day incubation in the osteogenic differentiation media, the integrin diffusion coefficients increased by an order of magnitude, and the integrin dynamics became indistinguishable from that measured on the surface of terminally differentiated human osteoblasts. Furthermore, microfilaments in BMPCs consisted of atypically thick bundles of stress fibers that were responsible for restricting the integrin lateral mobility. Studies using laser optical tweezers showed that, unlike fully differentiated osteoblasts, the BMPC cytoskeleton is weakly associated with its cell membrane. Based on these findings, it appears likely that the altered integrin dynamics is correlated with BMPC differentiation and that the integrin lateral mobility is restricted by direct links to microfilaments.
Collapse
Affiliation(s)
- Hongfeng Chen
- Department of Bioengineering, University of Illinois, Chicago, Illinois 60607, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Recent evidence shows that transplantation of neural stem/precursor cells may protect the central nervous system from inflammatory damage through a 'bystander' mechanism that is alternative to cell replacement. This novel mechanism, which might improve the success of transplantation procedures, is exerted by undifferentiated neural stem cells, the functional characteristics of which are regulated by important stem cell regulators released by CNS-resident and blood-borne inflammatory cells. Here, we discuss this alternative bystander mechanism in the context of the atypical ectopic perivascular niche. We propose that it is the most challenging example of reciprocal therapeutic crosstalk between the inflamed CNS and systemically transplanted neural stem cells.
Collapse
Affiliation(s)
- Gianvito Martino
- Neuroimmunology Unit, DIBIT, and Department of Neurology and Neurophysiology, San Raffaele Scientific Institute, via Olgettina 58, 20132, Milano, Italy.
| | | |
Collapse
|
23
|
Campos LS. Beta1 integrins and neural stem cells: making sense of the extracellular environment. Bioessays 2005; 27:698-707. [PMID: 15954093 DOI: 10.1002/bies.20256] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neural Stem Cells (NSC) are present in the developing and adult CNS. In both the embryonic and adult neurogenic regions, beta1 integrins may act as sensors for the changing extracellular matrix. Here we highlight the integrative functions that beta1 integrins may play in the "niche" by regulating NSC growth factor responsiveness in a timely and spatially controlled manner. beta1 integrins may provide NSC with the capacity to react to a dynamic "niche", and to respond adequately by either remaining as stem cells or by differentiating and migrating away to shape the developing cortex.
Collapse
|
24
|
Muramatsu D, Sato Y, Hishiyama S, Miyamoto Y, Hisatsune T. Transplantation of GABAergic neurons into adult mouse neocortex. Exp Neurol 2005; 194:1-11. [PMID: 15899239 DOI: 10.1016/j.expneurol.2005.01.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Revised: 12/17/2004] [Accepted: 01/11/2005] [Indexed: 11/25/2022]
Abstract
GABAergic neurons in the neocortex contribute to various brain functions by regulating cortical pyramidal neurons. A deficiency of GABAergic neurons in the neocortex leads to the dysregulation of cortical neuronal circuits, but this can be overcome by cell transplantation, which provides a practical approach to repair damaged neuronal circuits. Here, we focused on the transplantation of committed neuronal progenitor cells. Because neuronal differentiation is considerably suppressed in the adult neocortex, we transfected proneural bHLH transcription factors into neural precursor cells to commit them to a neuronal lineage prior to the cell transplantation. We show that ventral neural stem cells transfected with Ngn1 are integrated as GABAergic neurons within a few days of transplantation into the adult mouse neocortex. These results demonstrate that the transplantation of committed neuronal progenitor cells is an effective method for brain repair.
Collapse
Affiliation(s)
- Dai Muramatsu
- Department of Integrated Biosciences, University of Tokyo, Bioscience Building 402, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | | | | | | | | |
Collapse
|
25
|
Mutoh T, Hamada S, Senzaki K, Murata Y, Yagi T. Cadherin-related neuronal receptor 1 (CNR1) has cell adhesion activity with β1 integrin mediated through the RGD site of CNR1. Exp Cell Res 2004; 294:494-508. [PMID: 15023537 DOI: 10.1016/j.yexcr.2003.11.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2003] [Revised: 10/17/2003] [Indexed: 01/22/2023]
Abstract
Cadherin-related neuronal receptor (CNR) proteins are a diverse set of synaptic protocadherins, but little is known about its adhesive properties. We found that overexpressed CNR1 protein localized on the cell surface of HEK293T cells and increased the calcium-dependent cell aggregation potential. However, we could not detect the strong homophilic binding activity of CNR1 EC-Fc fusion protein in vitro. Parental HEK293T cells adhered to Arg-Gly-Asp (RGD) motif of EC1 domain of CNR1-Fc fusion protein. The fusion protein that the Asp73 of EC1 point-mutated to Glu (RGE-Fc) lost the adhesive activity. The adhesion activity of HEK293T cells to CNR1 EC-Fc fusion protein was completely blocked by inhibitors of integrins, including RGDS peptide and anti-beta1 integrin antibodies. The increased cell-aggregative property of CNR1 transfectants was also blocked by RGDS peptides. At cell-cell junctions of the CNR1 transfectants, co-localization between CNR1 and HEK293T endogenous beta1 integrin was observed. Furthermore, the spatiotemporal expression patterns of CNR and beta1 integrin nearly overlapped in the molecular layer of the developing mouse cerebellum in the main stage of synaptogenesis. These results indicate that CNR1 has a heterophilic, calcium-dependent cell adhesion activity with the beta1 integrin subfamily, and raise the possibility of CNR-beta1 integrin association in synaptogenesis.
Collapse
Affiliation(s)
- Tetsuji Mutoh
- KOKORO Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita 565-0871, Japan.
| | | | | | | | | |
Collapse
|