1
|
Ortega JA, Soares de Aguiar GP, Chandravanshi P, Levy N, Engel E, Álvarez Z. Exploring the properties and potential of the neural extracellular matrix for next-generation regenerative therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1962. [PMID: 38723788 DOI: 10.1002/wnan.1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024]
Abstract
The extracellular matrix (ECM) is a dynamic and complex network of proteins and molecules that surrounds cells and tissues in the nervous system and orchestrates a myriad of biological functions. This review carefully examines the diverse interactions between cells and the ECM, as well as the transformative chemical and physical changes that the ECM undergoes during neural development, aging, and disease. These transformations play a pivotal role in shaping tissue morphogenesis and neural activity, thereby influencing the functionality of the central nervous system (CNS). In our comprehensive review, we describe the diverse behaviors of the CNS ECM in different physiological and pathological scenarios and explore the unique properties that make ECM-based strategies attractive for CNS repair and regeneration. Addressing the challenges of scalability, variability, and integration with host tissues, we review how advanced natural, synthetic, and combinatorial matrix approaches enhance biocompatibility, mechanical properties, and functional recovery. Overall, this review highlights the potential of decellularized ECM as a powerful tool for CNS modeling and regenerative purposes and sets the stage for future research in this exciting field. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- J Alberto Ortega
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Gisele P Soares de Aguiar
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Palash Chandravanshi
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Natacha Levy
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Department of Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Zaida Álvarez
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
2
|
Mubuchi A, Takechi M, Nishio S, Matsuda T, Itoh Y, Sato C, Kitajima K, Kitagawa H, Miyata S. Assembly of neuron- and radial glial-cell-derived extracellular matrix molecules promotes radial migration of developing cortical neurons. eLife 2024; 12:RP92342. [PMID: 38512724 PMCID: PMC10957175 DOI: 10.7554/elife.92342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024] Open
Abstract
Radial neuronal migration is a key neurodevelopmental event for proper cortical laminar organization. The multipolar-to-bipolar transition, a critical step in establishing neuronal polarity during radial migration, occurs in the subplate/intermediate zone (SP/IZ), a distinct region of the embryonic cerebral cortex. It has been known that the extracellular matrix (ECM) molecules are enriched in the SP/IZ. However, the molecular constitution and functions of the ECM formed in this region remain poorly understood. Here, we identified neurocan (NCAN) as a major chondroitin sulfate proteoglycan in the mouse SP/IZ. NCAN binds to both radial glial-cell-derived tenascin-C (TNC) and hyaluronan (HA), a large linear polysaccharide, forming a ternary complex of NCAN, TNC, and HA in the SP/IZ. Developing cortical neurons make contact with the ternary complex during migration. The enzymatic or genetic disruption of the ternary complex impairs radial migration by suppressing the multipolar-to-bipolar transition. Furthermore, both TNC and NCAN promoted the morphological maturation of cortical neurons in vitro. The present results provide evidence for the cooperative role of neuron- and radial glial-cell-derived ECM molecules in cortical development.
Collapse
Affiliation(s)
- Ayumu Mubuchi
- Graduate School of Agriculture, Tokyo University of Agriculture and TechnologyFuchuJapan
| | - Mina Takechi
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
| | - Shunsuke Nishio
- Faculty of Food and Agricultural Sciences, Fukushima UniversityFukushimaJapan
| | - Tsukasa Matsuda
- Faculty of Food and Agricultural Sciences, Fukushima UniversityFukushimaJapan
| | - Yoshifumi Itoh
- Kennedy Institute of Rheumatology, University of OxfordOxfordUnited Kingdom
| | - Chihiro Sato
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Bioscience and Biotechnology Center, Nagoya UniversityNagoyaJapan
- Institute for Glyco-core Research, Nagoya UniversityNagoyaJapan
| | - Ken Kitajima
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Bioscience and Biotechnology Center, Nagoya UniversityNagoyaJapan
- Institute for Glyco-core Research, Nagoya UniversityNagoyaJapan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical UniversityKobeJapan
| | - Shinji Miyata
- Graduate School of Agriculture, Tokyo University of Agriculture and TechnologyFuchuJapan
| |
Collapse
|
3
|
Fu Z, Zhu G, Luo C, Chen Z, Dou Z, Chen Y, Zhong C, Su S, Liu F. Matricellular protein tenascin C: Implications in glioma progression, gliomagenesis, and treatment. Front Oncol 2022; 12:971462. [PMID: 36033448 PMCID: PMC9413079 DOI: 10.3389/fonc.2022.971462] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
Matricellular proteins are nonstructural extracellular matrix components that are expressed at low levels in normal adult tissues and are upregulated during development or under pathological conditions. Tenascin C (TNC), a matricellular protein, is a hexameric and multimodular glycoprotein with different molecular forms that is produced by alternative splicing and post-translational modifications. Malignant gliomas are the most common and aggressive primary brain cancer of the central nervous system. Despite continued advances in multimodal therapy, the prognosis of gliomas remains poor. The main reasons for such poor outcomes are the heterogeneity and adaptability caused by the tumor microenvironment and glioma stem cells. It has been shown that TNC is present in the glioma microenvironment and glioma stem cell niches, and that it promotes malignant properties, such as neovascularization, proliferation, invasiveness, and immunomodulation. TNC is abundantly expressed in neural stem cell niches and plays a role in neurogenesis. Notably, there is increasing evidence showing that neural stem cells in the subventricular zone may be the cells of origin of gliomas. Here, we review the evidence regarding the role of TNC in glioma progression, propose a potential association between TNC and gliomagenesis, and summarize its clinical applications. Collectively, TNC is an appealing focus for advancing our understanding of gliomas.
Collapse
Affiliation(s)
- Zaixiang Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ganggui Zhu
- Department of Neurosurgery, Hangzhou First People’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chao Luo
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Zihang Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhangqi Dou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yike Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chen Zhong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Su
- Department of Neurosurgery, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Fuyi Liu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Fuyi Liu,
| |
Collapse
|
4
|
The expression of tenascin-C in neural stem/progenitor cells is stimulated by the growth factors EGF and FGF-2, but not by TGFβ1. Cell Tissue Res 2021; 385:659-674. [PMID: 34309729 PMCID: PMC8526465 DOI: 10.1007/s00441-021-03508-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 07/10/2021] [Indexed: 12/26/2022]
Abstract
Neural stem/progenitor cells (NSPCs) rely on internal and external cues determining their lineage decisions during brain development. The progenitor cells of the embryonic mammalian forebrain reside in the ventricular and subventricular zones of the lateral ventricles, where they proliferate, generate neurons and glial cells, and respond to external cues like growth factors. The extracellular matrix (ECM) surrounds NSPCs and influences the cell fate by providing mechanical scaffold, trophic support, and instructive signals. The ECM molecule tenascin-C (Tnc) is expressed in the proliferative zones of the developing forebrain and involved in the proliferation and maturation of NSPCs. Here, we analyzed the regulation of the Tnc gene expression by NSPCs cultivated under the influence of different growth factors. We observed that the epidermal growth factor (EGF) and the fibroblast growth factor (FGF)-2 strongly increased the expression of Tnc, whereas the transforming growth factor (TGF)β 1 had no effect on Tnc gene expression, in contrast to previous findings in cell cultures of neural and non-neural origin. The stimulation of the Tnc gene expression induced by EGF or FGF-2 was reversible and seen in constantly treated as well as short term stimulated NSPC cultures. The activation depended on the presence of the respective receptors, which was slightly different in cortical and striatal NSPC cultures. Our results confirm the influence of extracellular stimuli regulating the expression of factors that form a niche for NSPCs during embryonic forebrain development.
Collapse
|
5
|
Tucić M, Stamenković V, Andjus P. The Extracellular Matrix Glycoprotein Tenascin C and Adult Neurogenesis. Front Cell Dev Biol 2021; 9:674199. [PMID: 33996833 PMCID: PMC8117239 DOI: 10.3389/fcell.2021.674199] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Tenascin C (TnC) is a glycoprotein highly expressed in the extracellular matrix (ECM) during development and in the adult central nervous system (CNS) in regions of active neurogenesis, where neuron development is a tightly regulated process orchestrated by extracellular matrix components. In addition, newborn cells also communicate with glial cells, astrocytes and microglia, indicating the importance of signal integration in adult neurogenesis. Although TnC has been recognized as an important molecule in the regulation of cell proliferation and migration, complete regulatory pathways still need to be elucidated. In this review we discuss the formation of new neurons in the adult hippocampus and the olfactory system with specific reference to TnC and its regulating functions in this process. Better understanding of the ECM signaling in the niche of the CNS will have significant implications for regenerative therapies.
Collapse
Affiliation(s)
- Milena Tucić
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vera Stamenković
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Pavle Andjus
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
6
|
Bijelić D, Adžić M, Perić M, Jakovčevski I, Förster E, Schachner M, Andjus PR. Different Functions of Recombinantly Expressed Domains of Tenascin-C in Glial Scar Formation. Front Immunol 2021; 11:624612. [PMID: 33679718 PMCID: PMC7934619 DOI: 10.3389/fimmu.2020.624612] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular matrix glycoprotein tenascin-C (TnC) is highly expressed in vertebrates during embryonic development and thereafter transiently in tissue niches undergoing extensive remodeling during regeneration after injury. TnC's different functions can be attributed to its multimodular structure represented by distinct domains and alternatively spliced isoforms. Upon central nervous system injury, TnC is upregulated and secreted into the extracellular matrix mainly by astrocytes. The goal of the present study was to elucidate the role of different TnC domains in events that take place after spinal cord injury (SCI). Astrocyte cultures prepared from TnC-deficient (TnC-/-) and wild-type (TnC+/+) mice were scratched and treated with different recombinantly generated TnC fragments. Gap closure, cell proliferation and expression of GFAP and cytokines were determined in these cultures. Gap closure in vitro was found to be delayed by TnC fragments, an effect mainly mediated by decreasing proliferation of astrocytes. The most potent effects were observed with fragments FnD, FnA and their combination. TnC-/- astrocyte cultures exhibited higher GFAP protein and mRNA expression levels, regardless of the type of fragment used for treatment. Application of TnC fragments induced also pro-inflammatory cytokine production by astrocytes in vitro. In vivo, however, the addition of FnD or Fn(D+A) led to a difference between the two genotypes, with higher levels of GFAP expression in TnC+/+ mice. FnD treatment of injured TnC-/- mice increased the density of activated microglia/macrophages in the injury region, while overall cell proliferation in the injury site was not affected. We suggest that altogether these results may explain how the reaction of astrocytes is delayed while their localization is restricted to the border of the injury site to allow microglia/macrophages to form a lesion core during the first stages of glial scar formation, as mediated by TnC and, in particular, the alternatively spliced FnD domain.
Collapse
Affiliation(s)
- Dunja Bijelić
- Centre for Laser Microscopy, Faculty of Biology, Institute of Physiology and Biochemistry "Jean Giaja", University of Belgrade, Belgrade, Serbia
| | - Marija Adžić
- Centre for Laser Microscopy, Faculty of Biology, Institute of Physiology and Biochemistry "Jean Giaja", University of Belgrade, Belgrade, Serbia
| | - Mina Perić
- Centre for Laser Microscopy, Faculty of Biology, Institute of Physiology and Biochemistry "Jean Giaja", University of Belgrade, Belgrade, Serbia
| | - Igor Jakovčevski
- Institut für Neuroanatomie und Molekulare Hirnforschung, Ruhr-Universität Bochum, Bochum, Germany
| | - Eckart Förster
- Institut für Neuroanatomie und Molekulare Hirnforschung, Ruhr-Universität Bochum, Bochum, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Pavle R Andjus
- Centre for Laser Microscopy, Faculty of Biology, Institute of Physiology and Biochemistry "Jean Giaja", University of Belgrade, Belgrade, Serbia
| |
Collapse
|
7
|
de Jong JM, Wang P, Oomkens M, Baron W. Remodeling of the interstitial extracellular matrix in white matter multiple sclerosis lesions: Implications for remyelination (failure). J Neurosci Res 2020; 98:1370-1397. [PMID: 31965607 DOI: 10.1002/jnr.24582] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/29/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) provides protection, rigidity, and structure toward cells. It consists, among others, of a wide variety of glycoproteins and proteoglycans, which act together to produce a complex and dynamic environment, most relevant in transmembrane events. In the brain, the ECM occupies a notable proportion of its volume and maintains the homeostasis of central nervous system (CNS). In addition, remodeling of the ECM, that is transient changes in ECM proteins regulated by matrix metalloproteinases (MMPs), is an important process that modulates cell behavior upon injury, thereby facilitating recovery. Failure of ECM remodeling plays an important role in the pathogenesis of multiple sclerosis (MS), a neurodegenerative demyelinating disease of the CNS with an inflammatory response against protective myelin sheaths that surround axons. Remyelination of denuded axons improves the neuropathological conditions of MS, but this regeneration process fails over time, leading to chronic disease progression. In this review, we uncover abnormal ECM remodeling in MS lesions by discussing ECM remodeling in experimental demyelination models, that is when remyelination is successful, and compare alterations in ECM components to the ECM composition and MMP expression in the parenchyma of demyelinated MS lesions, that is when remyelination fails. Inter- and intralesional differences in ECM remodeling in the distinct white matter MS lesions are discussed in terms of consequences for oligodendrocyte behavior and remyelination (failure). Hence, the review will aid to understand how abnormal ECM remodeling contributes to remyelination failure in MS lesions and assists in developing therapeutic strategies to promote remyelination.
Collapse
Affiliation(s)
- Jody M de Jong
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peng Wang
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michelle Oomkens
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wia Baron
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
8
|
Ha S, Jeong SH, Yi K, Chu JJM, Kim S, Kim EK, Yu SW. Autophagy Mediates Astrogenesis in Adult Hippocampal Neural Stem Cells. Exp Neurobiol 2019; 28:229-246. [PMID: 31138991 PMCID: PMC6526118 DOI: 10.5607/en.2019.28.2.229] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 12/14/2022] Open
Abstract
Neural stem cells (NSCs) have the ability to self-renew and differentiate into neurons, oligodendrocytes, and astrocytes. Highly dynamic nature of NSC differentiation requires the intimate involvement of catabolic processes such as autophagy. Autophagy is a major intracellular degradation pathway necessary for cellular homeostasis and remodeling. Autophagy is important for mammalian development and its role in neurogenesis has recently drawn much attention. However, little is known about how autophagy is associated with differentiation of NSCs into other neural lineages. Here, we report that autophagy plays a critical role in differentiation of adult rat hippocampal neural stem (HCN) cells into astrocytes. During differentiation, autophagy flux peaked at early time points, and remained high. Pharmacological or genetic suppression of autophagy by stable knockdown of Atg7, LC3 or CRISPR-Cas9-mediated knockout (KO) of p62 impaired astrogenesis, while reintroduction of p62 recovered astrogenesis in p62 KO HCN cells. Taken together, our findings suggest that autophagy plays a key role in astrogenesis in adult NSCs.
Collapse
Affiliation(s)
- Shinwon Ha
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Seol-Hwa Jeong
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Kyungrim Yi
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Jamie Jeong-Min Chu
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Seolsong Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Eun-Kyoung Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea.,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Seong-Woon Yu
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea.,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
9
|
May M, Denecke B, Schroeder T, Götz M, Faissner A. Cell tracking in vitro reveals that the extracellular matrix glycoprotein Tenascin-C modulates cell cycle length and differentiation in neural stem/progenitor cells of the developing mouse spinal cord. Biol Open 2018; 7:7/7/bio027730. [PMID: 30045859 PMCID: PMC6078350 DOI: 10.1242/bio.027730] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Generation of astrocytes during the development of the mammalian spinal cord is poorly understood. Previously, we have shown that the glycoprotein of the extracellular matrix (ECM) tenascin-C (Tnc) modulates the expression territories of the patterning genes Nkx6.1 and Nkx2.2 in the developing ventral spinal cord, tunes the responsiveness of neural stem/progenitor cells towards the cytokines FGF2 and EGF and thereby promotes astrocyte maturation. In order to obtain further mechanistic insight into these processes, we have compared embryonic day-15 spinal cord neural progenitor cells (NPCs) from wild-type and Tnc knockout mice using continuous single-cell live imaging and cell lineage analysis in vitroTnc knockout cells displayed a significantly reduced rate of cell division both in response to FGF2 and EGF. When individual clones of dividing cells were investigated with regard to their cell lineage trees using the tTt tracking software, it appeared that the cell cycle length in response to growth factors was reduced in the knockout. Furthermore, when Tnc knockout NPCs were induced to differentiate by the removal of FGF2 and EGF glial differentiation was enhanced. We conclude that the constituent of the stem cell niche Tnc contributes to preserve stemness of NPCs.
Collapse
Affiliation(s)
- Marcus May
- Department for Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Bernd Denecke
- Aachen Interdisciplinary Center for Clinical Research, Faculty of Medicine, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zürich, 4058 Basel, Switzerland
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg/Martinsried, Germany.,Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg/Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians University Munich, 81377 Munich, Germany
| | - Andreas Faissner
- Department for Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| |
Collapse
|
10
|
The Tenascin-C-Derived Peptide VSWRAPTA Promotes Neuronal Branching Via Transcellular Activation of the Focal Adhesion Kinase (FAK) and the ERK1/2 Signaling Pathway In Vitro. Mol Neurobiol 2018; 56:632-647. [DOI: 10.1007/s12035-018-1108-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/03/2018] [Indexed: 12/27/2022]
|
11
|
Role of Matricellular Proteins in Disorders of the Central Nervous System. Neurochem Res 2016; 42:858-875. [DOI: 10.1007/s11064-016-2088-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022]
|
12
|
Stanic K, Saldivia N, Förstera B, Torrejón M, Montecinos H, Caprile T. Expression Patterns of Extracellular Matrix Proteins during Posterior Commissure Development. Front Neuroanat 2016; 10:89. [PMID: 27733818 PMCID: PMC5039192 DOI: 10.3389/fnana.2016.00089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/12/2016] [Indexed: 11/13/2022] Open
Abstract
Extracellular matrix (ECM) molecules are pivotal for central nervous system (CNS) development, facilitating cell migration, axonal growth, myelination, dendritic spine formation, and synaptic plasticity, among other processes. During axon guidance, the ECM not only acts as a permissive or non-permissive substrate for navigating axons, but also modulates the effects of classical guidance cues, such as netrin or Eph/ephrin family members. Despite being highly important, little is known about the expression of ECM molecules during CNS development. Therefore, this study assessed the molecular expression patterns of tenascin, HNK-1, laminin, fibronectin, perlecan, decorin, and osteopontin along chick embryo prosomere 1 during posterior commissure development. The posterior commissure is the first transversal axonal tract of the embryonic vertebrate brain. Located in the dorso-caudal portion of prosomere 1, posterior commissure axons primarily arise from the neurons of basal pretectal nuclei that run dorsally to the roof plate midline, where some turn toward the ipsilateral side. Expressional analysis of ECM molecules in this area these revealed to be highly arranged, and molecule interactions with axon fascicles suggested involvement in processes other than structural support. In particular, tenascin and the HNK-1 epitope extended in ventro-dorsal columns and enclosed axons during navigation to the roof plate. Laminin and osteopontin were expressed in the midline, very close to axons that at this point must decide between extending to the contralateral side or turning to the ipsilateral side. Finally, fibronectin, decorin, and perlecan appeared unrelated to axonal pathfinding in this region and were instead restricted to the external limiting membrane. In summary, the present report provides evidence for an intricate expression of different extracellular molecules that may cooperate in guiding posterior commissure axons.
Collapse
Affiliation(s)
- Karen Stanic
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Natalia Saldivia
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Benjamín Förstera
- Department of Physiology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Marcela Torrejón
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Hernán Montecinos
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Teresa Caprile
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| |
Collapse
|
13
|
Faissner A. REVIEW ■ : Glial Derived Extracellular Matrix Components: Important Roles in Axon Growth and Guidance. Neuroscientist 2016. [DOI: 10.1177/107385849700300610] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Axon growth and guidance, and the correct recognition of distant targets by growth cones rank among the most spectacular achievements of the developing nervous system. The establishment and reformation of adequate networks and the plasticity of synaptic connections are vital for the function and the restoration of the nervous system under conditions of health and disease. Therefore, considerable efforts have been devoted to the elucidation of the molecular and cellular bases of the establishment of interneuronal con nections. It is well established that interactions between neurons and astrocytes are of regulatory importance in this context. Thus, astroglia guides migrating neurons and advancing growth cones to their destination. On the other hand, astrocytes design transient boundaries that deflect axons and segregate groups of neurons, and form scars involved in the inhibition of axonal regeneration after lesion. This duplicity of astroglia is presumably mediated by various gene families. Among these, extracellular matrix (ECM) con stituents seem particularly suited to embody and mediate the ambivalence of astrocytes because these compounds appear to exert either conducive or inhibitory/repulsive effects depending on interacting cell types and conditions. Furthermore, ECM constituents are upregulated by astrocytes upon lesion and con tribute to the construction of glial scars. This review focuses on this class of compounds and their possible functions in the wiring of neural networks. NEUROSCIENTIST 3:371-380, 1997
Collapse
|
14
|
Abstract
Tenascin-C is a large, multimodular, extracellular matrix glycoprotein that exhibits a very restricted pattern of expression but an enormously diverse range of functions. Here, we discuss the importance of deciphering the expression pattern of, and effects mediated by, different forms of this molecule in order to fully understand tenascin-C biology. We focus on both post transcriptional and post translational events such as splicing, glycosylation, assembly into a 3D matrix and proteolytic cleavage, highlighting how these modifications are key to defining tenascin-C function.
Collapse
Key Words
- AD1/AD2, additional domain 1/ additional domain 2
- ADAMTS, a disintegrin and metalloproteinase with thrombospondin motifs
- ASMCs, aortic smooth muscle cells
- BDNF, brain derived neurotrophic factor
- BHKs, baby hamster kidney cells
- BMP, bone morphogenetic protein
- CA19–9, carbohydrate antigen 19–9
- CALEB, chicken acidic leucine-rich EGF-like domain containing brain protein
- CEA, carcinoembryonic antigen
- CNS, central nervous system
- CRC, colorectal carcinomas
- CTGF, connective tissue growth factor
- DCIS, ductal carcinoma in-situ
- ECM, extracellular matrix
- EDA-FN, extra domain A containing fibronectin
- EDB-FN, extra domain B containing fibronectin
- EGF-L, epidermal growth factor-like
- EGF-R, epidermal growth factor receptor
- ELISPOT, enzyme-linked immunospot assay
- FBG, fibrinogen-like globe
- FGF2, fibroblast growth factor 2
- FGF4, fibroblast growth factor 4
- FN, fibronectin
- FNIII, fibronectin type III-like repeat
- GMEM, glioma-mesenchymal extracellular matrix antigen
- GPI, glycosylphosphatidylinositol
- HB-EGF, heparin-binding EGF-like growth factor
- HCEs, immortalized human corneal epithelial cell line
- HGF, hepatocyte growth factor
- HNK-1, human natural killer-1
- HSPGs, heparan sulfate proteoglycans
- HUVECs, human umbilical vein endothelial cells
- ICC, immunocytochemistry
- IF, immunofluorescence
- IFNγ, interferon gamma
- IGF, insulin-like growth factor
- IGF-BP, insulin-like growth factor-binding protein
- IHC, immunohistochemistry
- IL, interleukin
- ISH, in situ hybridization
- LPS, lipopolysaccharide
- MMP, matrix metalloproteinase
- MPNSTs, malignant peripheral nerve sheath tumors
- Mr, molecular mass
- NB, northern blot
- NF-kB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NK, natural killer cells
- NSCLC, non-small cell lung carcinoma
- NSCs, neural stem cells
- NT, neurotrophin
- PAMPs, pathogen-associated molecular patterns
- PDGF, platelet derived growth factor
- PDGF-Rβ, platelet derived growth factor receptor β
- PIGF, phosphatidylinositol-glycan biosynthesis class F protein
- PLCγ, phospholipase-C gamma
- PNS, peripheral nervous system
- PTPRζ1, receptor-type tyrosine-protein phosphatase zeta
- RA, rheumatoid arthritis
- RCC, renal cell carcinoma
- RD, rhabdomyosarcoma
- RGD, arginylglycylaspartic acid
- RT-PCR, real-time polymerase chain reaction
- SB, Southern blot
- SCC, squamous cell carcinoma
- SMCs, smooth muscle cells
- SVZ, sub-ventricular zone
- TA, tenascin assembly domain
- TGFβ, transforming growth factor β
- TIMP, tissue inhibitor of metalloproteinases
- TLR4, toll-like receptor 4
- TNFα, tumor necrosis factor α
- TSS, transcription start site
- UBC, urothelial bladder cancer
- UCC, urothelial cell carcinoma
- VEGF, vascular endothelial growth factor
- VSMCs, vascular smooth muscle cells
- VZ, ventricular zone
- WB, immunoblot/ western blot
- bFGF, basic fibroblast growth factor
- biosynthesis
- c, charged
- cancer
- ccRCC, clear cell renal cell carcinoma
- chRCC, chromophobe-primary renal cell carcinoma
- development
- glycosylation
- mAb, monoclonal antibody
- matrix assembly
- mitogen-activated protein kinase, MAPK
- pHo, extracellular pH
- pRCC, papillary renal cell carcinoma
- proteolytic cleavage
- siRNA, small interfering RNA
- splicing
- tenascin-C
- therapeutics
- transcription
Collapse
Affiliation(s)
- Sean P Giblin
- a Nuffield Department of Orthopaedics; Rheumatology and Musculoskeletal Sciences ; Kennedy Institute of Rheumatology; University of Oxford ; Oxford , UK
| | | |
Collapse
|
15
|
Lhx2 regulates the timing of β-catenin-dependent cortical neurogenesis. Proc Natl Acad Sci U S A 2015; 112:12199-204. [PMID: 26371318 DOI: 10.1073/pnas.1507145112] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The timing of cortical neurogenesis has a major effect on the size and organization of the mature cortex. The deletion of the LIM-homeodomain transcription factor Lhx2 in cortical progenitors by Nestin-cre leads to a dramatically smaller cortex. Here we report that Lhx2 regulates the cortex size by maintaining the cortical progenitor proliferation and delaying the initiation of neurogenesis. The loss of Lhx2 in cortical progenitors results in precocious radial glia differentiation and a temporal shift of cortical neurogenesis. We further investigated the underlying mechanisms at play and demonstrated that in the absence of Lhx2, the Wnt/β-catenin pathway failed to maintain progenitor proliferation. We developed and applied a mathematical model that reveals how precocious neurogenesis affected cortical surface and thickness. Thus, we concluded that Lhx2 is required for β-catenin function in maintaining cortical progenitor proliferation and controls the timing of cortical neurogenesis.
Collapse
|
16
|
Reinhard J, Joachim SC, Faissner A. Extracellular matrix remodeling during retinal development. Exp Eye Res 2015; 133:132-40. [DOI: 10.1016/j.exer.2014.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 10/25/2022]
|
17
|
Gaudet AD, Popovich PG. Extracellular matrix regulation of inflammation in the healthy and injured spinal cord. Exp Neurol 2014; 258:24-34. [PMID: 25017885 DOI: 10.1016/j.expneurol.2013.11.020] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 02/06/2023]
Abstract
Throughout the body, the extracellular matrix (ECM) provides structure and organization to tissues and also helps regulate cell migration and intercellular communication. In the injured spinal cord (or brain), changes in the composition and structure of the ECM undoubtedly contribute to regeneration failure. Less appreciated is how the native and injured ECM influences intraspinal inflammation and, conversely, how neuroinflammation affects the synthesis and deposition of ECM after CNS injury. In all tissues, inflammation can be initiated and propagated by ECM disruption. Molecules of ECM newly liberated by injury or inflammation include hyaluronan fragments, tenascins, and sulfated proteoglycans. These act as "damage-associated molecular patterns" or "alarmins", i.e., endogenous proteins that trigger and subsequently amplify inflammation. Activated inflammatory cells, in turn, further damage the ECM by releasing degradative enzymes including matrix metalloproteinases (MMPs). After spinal cord injury (SCI), destabilization or alteration of the structural and chemical compositions of the ECM affects migration, communication, and survival of all cells - neural and non-neural - that are critical for spinal cord repair. By stabilizing ECM structure or modifying their ability to trigger the degradative effects of inflammation, it may be possible to create an environment that is more conducive to tissue repair and axon plasticity after SCI.
Collapse
Affiliation(s)
- Andrew D Gaudet
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, College of Medicine, The Ohio State University, 670 Biomedical Research Tower, 460 West 12th Ave., Columbus, OH 43210, USA.
| | - Phillip G Popovich
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, College of Medicine, The Ohio State University, 670 Biomedical Research Tower, 460 West 12th Ave., Columbus, OH 43210, USA.
| |
Collapse
|
18
|
Jakovcevski I, Miljkovic D, Schachner M, Andjus PR. Tenascins and inflammation in disorders of the nervous system. Amino Acids 2012; 44:1115-27. [DOI: 10.1007/s00726-012-1446-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 12/10/2012] [Indexed: 12/20/2022]
|
19
|
Kazanis I, ffrench-Constant C. Extracellular matrix and the neural stem cell niche. Dev Neurobiol 2012; 71:1006-17. [PMID: 21898854 DOI: 10.1002/dneu.20970] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Basal lamina is present in many stem cell niches, but we still have a poor understanding of the role of this and other extracellular matrix (ECM) components. Here, we review current knowledge regarding ECM expression and function in the neural stem cell niche, focusing on the subependymal zone of the adult CNS. An increasing complexity of ECM molecules has been described, and a number of receptors expressed on the stem cells identified. Experiments perturbing the niche using genetics or cytotoxic ablation of the rapidly dividing precursors, or using explant culture models to examine specific growth factors, have been influential in showing how changes in these ECM receptors might regulate neural stem cell behavior. However the role of changes in the matrix itself remains to be determined. The answers will be important, as they will point to the molecules required to engineer niches ex-vivo so as to provide tools for regenerative neuroscience.
Collapse
Affiliation(s)
- Ilias Kazanis
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
20
|
Wiese S, Karus M, Faissner A. Astrocytes as a source for extracellular matrix molecules and cytokines. Front Pharmacol 2012; 3:120. [PMID: 22740833 PMCID: PMC3382726 DOI: 10.3389/fphar.2012.00120] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 06/06/2012] [Indexed: 12/19/2022] Open
Abstract
Research of the past 25 years has shown that astrocytes do more than participating and building up the blood-brain barrier and detoxify the active synapse by reuptake of neurotransmitters and ions. Indeed, astrocytes express neurotransmitter receptors and, as a consequence, respond to stimuli. Within the tripartite synapse, the astrocytes owe more and more importance. Besides the functional aspects the differentiation of astrocytes has gained a more intensive focus. Deeper knowledge of the differentiation processes during development of the central nervous system might help explaining and even help treating neurological diseases like Alzheimer’s disease, Amyotrophic lateral sclerosis, Parkinsons disease, and psychiatric disorders in which astrocytes have been shown to play a role. Specific differentiation of neural stem cells toward the astroglial lineage is performed as a multi-step process. Astrocytes and oligodendrocytes develop from a multipotent stem cell that prior to this has produced primarily neuronal precursor cells. This switch toward the more astroglial differentiation is regulated by a change in receptor composition on the cell surface and responsiveness to Fibroblast growth factor and Epidermal growth factor (EGF). The glial precursor cell is driven into the astroglial direction by signaling molecules like Ciliary neurotrophic factor, Bone Morphogenetic Proteins, and EGF. However, the early astrocytes influence their environment not only by releasing and responding to diverse soluble factors but also express a wide range of extracellular matrix (ECM) molecules, in particular proteoglycans of the lectican family and tenascins. Lately these ECM molecules have been shown to participate in glial development. In this regard, especially the matrix protein Tenascin C (Tnc) proved to be an important regulator of astrocyte precursor cell proliferation and migration during spinal cord development. Nevertheless, ECM molecules expressed by reactive astrocytes are also known to act mostly in an inhibitory fashion under pathophysiological conditions. Thus, we further summarize resent data concerning the role of chondroitin sulfate proteoglycans and Tnc under pathological conditions.
Collapse
Affiliation(s)
- Stefan Wiese
- Group for Molecular Cell Biology, Department for Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany
| | | | | |
Collapse
|
21
|
Shimamura M, Taniyama Y, Katsuragi N, Koibuchi N, Kyutoku M, Sato N, Allahtavakoli M, Wakayama K, Nakagami H, Morishita R. Role of central nervous system periostin in cerebral ischemia. Stroke 2012; 43:1108-14. [PMID: 22308244 DOI: 10.1161/strokeaha.111.636662] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND PURPOSE Although periostin, an extracellular matrix glycoprotein, plays pivotal roles in survival, migration, and regeneration in various cells, its expression and function in the brain are still unknown. Here, we investigated the expression and role of periostin in the ischemic brain. METHODS Expression of full-length periostin (periostin 1 [Pn1]) and its splicing variant lacking exon 17 (periostin 2 [Pn2]) was examined by real-time reverse transcription polymerase chain reaction (RT-PCR), Western blotting, and immunohistochemical staining in male C57BL6/J mice. The actions of periostin were examined in adult primary neuronal culture and in a transient middle cerebral artery occlusion (tMCAo) model. RESULTS Expression of Pn2, but not of Pn1, mRNA was markedly changed after tMCAo. Pn2 mRNA was decreased in the ischemic core at 3 hours after ischemia. At 24 hours, Pn2 mRNA was significantly increased in both the peri-ischemic and ischemic regions. Periostin was mainly observed in neurons in normal brain. However, neuronal expression of periostin was decreased temporarily in the ischemic region, but increased in astrocytes and around endothelial cells at 24 hours after tMCAo. Of importance, intracerebroventricular injection of Pn2 resulted in a significant reduction in infarct volume at 24 hours after tMCAo associated with phosphorylation of Akt. Also, the Pn2-treated mice survived longer until 1 week after tMCAo. Pn2 significantly inhibited neuronal death under hypoxia and stimulated neurite outgrowth. CONCLUSIONS Here, we demonstrated that periostin was expressed in the brain, and exogenous Pn2 exhibited neuroprotective effects and accelerated neurite outgrowth. Additional studies on periostin may provide new insights into the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Munehisa Shimamura
- Department of Clinical Gene Therapy, Osaka University, Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Roll L, Mittmann T, Eysel UT, Faissner A. The laser lesion of the mouse visual cortex as a model to study neural extracellular matrix remodeling during degeneration, regeneration and plasticity of the CNS. Cell Tissue Res 2012; 349:133-45. [DOI: 10.1007/s00441-011-1313-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/20/2011] [Indexed: 02/06/2023]
|
23
|
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
|
24
|
Yu YM, Cristofanilli M, Valiveti A, Ma L, Yoo M, Morellini F, Schachner M. The extracellular matrix glycoprotein tenascin-C promotes locomotor recovery after spinal cord injury in adult zebrafish. Neuroscience 2011; 183:238-50. [PMID: 21443931 DOI: 10.1016/j.neuroscience.2011.03.043] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 03/19/2011] [Accepted: 03/21/2011] [Indexed: 12/31/2022]
Abstract
Adult zebrafish, by virtue of exhibiting spontaneous recovery after spinal lesion, have evolved into a paradigmatic vertebrate model system to identify novel genes vital for successful regeneration after spinal cord injury. Due to a remarkable level of conservation between zebrafish and human genomes, such genes, once identified, could point to possibilities for addressing the multiple issues on how to deal with functional recovery after spinal cord injury in humans. In the current study, the extracellular matrix glycoprotein tenascin-C was studied in the zebrafish spinal cord injury model to assess the often disparate functions of this multidomain molecule under in vivo conditions. This in vivo study was deemed necessary since in vitro studies had shown discrepant functional effects on neurite outgrowth: tenascin-C inhibits neurite outgrowth when presented as a molecular barrier adjacent to a conducive substrate, but enhances neurite outgrowth when presented as a uniform substrate. Thus, our current study addresses the question as to which of these features prevails in vivo: whether tenascin-C reduces or enhances axonal regrowth after injury in a well accepted vertebrate model of spinal cord injury. We show upregulation of tenascin-C expression in regenerating neurons of the nucleus of median longitudinal fascicle (NMLF) in the brainstem and spinal motoneurons. Inhibition of tenascin-C expression by antisense oligonucleotide (morpholino) resulted in impaired locomotor recovery, reduced regrowth of axons from brainstem neurons and reduced synapse formation by the regrowing brainstem axons on spinal motoneurons, all vital indicators of regeneration. Our results thus point to an advantageous role of tenascin-C in promoting spinal cord regeneration, by promoting axonal regrowth and synapse formation in the spinal cord caudal to the lesion site after injury.
Collapse
Affiliation(s)
- Y-M Yu
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, 604 Allison Road, NJ 08854, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Guttery DS, Shaw JA, Lloyd K, Pringle JH, Walker RA. Expression of tenascin-C and its isoforms in the breast. Cancer Metastasis Rev 2011; 29:595-606. [PMID: 20814719 DOI: 10.1007/s10555-010-9249-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tenascin-C (TNC) is an extracellular matrix glycoprotein which is frequently up-regulated in a variety of pathological conditions including chronic inflammation and cancer. TNC has been implicated in the modulation of cell migration, proliferation, invasion and angiogenesis. Multiple isoforms of TNC can be generated through the alternative splicing of nine exons located in the fibronectin type III region of the molecule. The profile of isoforms expressed differs between cancers and normal breast, with the fully truncated TNC isoform being predominant in normal and benign tissues and higher molecular weight isoforms induced predominantly in cancer. The addition of extra domains within the fibronectin type III repeat domain greatly affects TNC function with multiple exon combinations available for splicing. Exons 14 and 16 are considered to be tumour-associated and have been shown to affect breast cell line invasion and growth in vitro to a greater extent than the full-length TNC isoform. This mini review will provide a summary of the literature to date regarding the expression of TNC isoforms in the breast and also discuss more recent developments in the field regarding exon AD1.
Collapse
Affiliation(s)
- David S Guttery
- Department of Cancer Studies and Molecular Medicine, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK.
| | | | | | | | | |
Collapse
|
26
|
Dobbertin A, Czvitkovich S, Theocharidis U, Garwood J, Andrews MR, Properzi F, Lin R, Fawcett JW, Faissner A. Analysis of combinatorial variability reveals selective accumulation of the fibronectin type III domains B and D of tenascin-C in injured brain. Exp Neurol 2010; 225:60-73. [PMID: 20451518 DOI: 10.1016/j.expneurol.2010.04.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 04/23/2010] [Accepted: 04/30/2010] [Indexed: 10/19/2022]
Abstract
Tenascin-C (Tnc) is a multimodular extracellular matrix glycoprotein that is markedly upregulated in CNS injuries where it is primarily secreted by reactive astrocytes. Different Tnc isoforms can be generated by the insertion of variable combinations of one to seven (in rats) alternatively spliced distinct fibronectin type III (FnIII) domains to the smallest variant. Each spliced FnIII repeat mediates specific actions on neurite outgrowth, neuron migration or adhesion. Hence, different Tnc isoforms might differentially influence CNS repair. We explored the expression pattern of Tnc variants after cortical lesions and after treatment of astrocytes with various cytokines. Using RT-PCR, we observed a strong upregulation of Tnc transcripts containing the spliced FnIII domains B or D in injured tissue at 2-4 days post-lesion (dpl). Looking at specific combinations, we showed a dramatic increase of Tnc isoforms harboring the neurite outgrowth-promoting BD repeat with both the B and D domains being adjacent to each other. Isoforms containing only the axon growth-stimulating spliced domain D were also dramatically enhanced after injury. Injury-induced increase of Tnc proteins comprising the domain D was confirmed by Western Blotting and immunostaining of cortical lesions. In contrast, the FnIII modules C and AD1 were weakly modulated after injury. The growth cone repulsive A1A2A4 domains were poorly expressed in normal and injured tissue but the smallest isoform, which is also repellant, was highly expressed after injury. Expression of the shortest Tnc isoform and of variants containing B, D or BD, was strongly upregulated in cultured astrocytes after TGFbeta1 treatment, suggesting that TGFbeta1 could mediate, at least in part, the injury-induced upregulation of these isoforms. We identified complex injury-induced differential regulations of Tnc isoforms that may well influence axonal regeneration and repair processes in the damaged CNS.
Collapse
Affiliation(s)
- Alexandre Dobbertin
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University of Bochum, 44780 Bochum, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Czopka T, Von Holst A, Schmidt G, Ffrench-Constant C, Faissner A. Tenascin C and tenascin R similarly prevent the formation of myelin membranes in a RhoA-dependent manner, but antagonistically regulate the expression of myelin basic protein via a separate pathway. Glia 2009; 57:1790-801. [DOI: 10.1002/glia.20891] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Abstract
The extracellular matrix molecule tenascin-C is highly expressed during embryonic development, tissue repair and in pathological situations such as chronic inflammation and cancer. Tenascin-C interacts with several other extracellular matrix molecules and cell-surface receptors, thus affecting tissue architecture, tissue resilience and cell responses. Tenascin-C modulates cell migration, proliferation and cellular signaling through induction of pro-inflammatory cytokines and oncogenic signaling molecules amongst other mechanisms. Given the causal role of inflammation in cancer progression, common mechanisms might be controlled by tenascin-C during both events. Drugs targeting the expression or function of tenascin-C or the tenascin-C protein itself are currently being developed and some drugs have already reached advanced clinical trials. This generates hope that increased knowledge about tenascin-C will further improve management of diseases with high tenascin-C expression such as chronic inflammation, heart failure, artheriosclerosis and cancer.
Collapse
|
29
|
Midwood KS, Orend G. The role of tenascin-C in tissue injury and tumorigenesis. J Cell Commun Signal 2009; 3:287-310. [PMID: 19838819 PMCID: PMC2778592 DOI: 10.1007/s12079-009-0075-1] [Citation(s) in RCA: 312] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 09/30/2009] [Indexed: 01/14/2023] Open
Abstract
The extracellular matrix molecule tenascin-C is highly expressed during embryonic development, tissue repair and in pathological situations such as chronic inflammation and cancer. Tenascin-C interacts with several other extracellular matrix molecules and cell-surface receptors, thus affecting tissue architecture, tissue resilience and cell responses. Tenascin-C modulates cell migration, proliferation and cellular signaling through induction of pro-inflammatory cytokines and oncogenic signaling molecules amongst other mechanisms. Given the causal role of inflammation in cancer progression, common mechanisms might be controlled by tenascin-C during both events. Drugs targeting the expression or function of tenascin-C or the tenascin-C protein itself are currently being developed and some drugs have already reached advanced clinical trials. This generates hope that increased knowledge about tenascin-C will further improve management of diseases with high tenascin-C expression such as chronic inflammation, heart failure, artheriosclerosis and cancer.
Collapse
Affiliation(s)
- Kim S. Midwood
- Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College of Science, Technology and Medicine, 65 Aspenlea Road, Hammersmith, London, W6 8LH UK
| | - Gertraud Orend
- Inserm U682, Strasbourg, 67200 France
- University of Strasbourg, UMR-S682, Strasbourg, 67081 France
- Department of Molecular Biology, CHRU Strasbourg, Strasbourg, 67200 France
| |
Collapse
|
30
|
Comparative screening of glial cell types reveals extracellular matrix that inhibits retinal axon growth in a chondroitinase ABC-resistant fashion. Glia 2009; 57:1420-38. [DOI: 10.1002/glia.20860] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
31
|
The glia-derived extracellular matrix glycoprotein tenascin-C promotes embryonic and postnatal retina axon outgrowth via the alternatively spliced fibronectin type III domain TNfnD. ACTA ACUST UNITED AC 2009; 4:271-83. [DOI: 10.1017/s1740925x09990020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Tenascin-C (Tnc) is an astrocytic multifunctional extracellular matrix (ECM) glycoprotein that potentially promotes or inhibits neurite outgrowth. To investigate its possible functions for retinal development, explants from embryonic day 18 (E18) rat retinas were cultivated on culture substrates composed of poly-d-lysine (PDL), or PDL additionally coated with Tnc or laminin (LN)-1, which significantly increased fiber length. When combined with LN, Tnc induced axon fasciculation that reduced the apparent number of outgrowing fibers. In order to circumscribe the stimulatory region, Tnc-derived fibronectin type III (TNfn) domains fused to the human Ig-Fc-fragment TNfnD6-Fc, TNfnBD-Fc, TNFnA1A2-Fc and TNfnA1D-Fc were studied. The fusion proteins TNfnBD-Fc and to a lesser degree TNfnA1D-Fc were stimulatory when compared with the Ig-Fc-fragment protein without insert. In contrast, the combination TNfnA1A2-Fc reduced fiber outgrowth beneath the values obtained for the Ig-Fc domain, indicating potential inhibitory properties. The monoclonal J1/tn2 antibody (clone 578) that is specific for domain TNfnD blocked the stimulatory properties of the TNfn-Fc fusions. When postnatal day 7 retinal ganglion cells were used rather that explants, Tnc and Tnc-derived proteins proved permissive for neurite outgrowth. The present study highlights a strong retinal axon growth-promoting activity of the Tnc domain TNfnD, which is modulated by neighboring domains.
Collapse
|
32
|
Cheng SM, Carr CE. Functional delay of myelination of auditory delay lines in the nucleus laminaris of the barn owl. Dev Neurobiol 2008; 67:1957-74. [PMID: 17918244 DOI: 10.1002/dneu.20541] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In the barn owl, maps of interaural time difference (ITD) are created in the nucleus laminaris (NL) by interdigitating axons that act as delay lines. Adult delay line axons are myelinated, and this myelination is timely, coinciding with the attainment of adult head size, and stable ITD cues. The proximal portions of the axons become myelinated in late embryonic life, but the delay line portions of the axon in NL remain unmyelinated until the first postnatal week. Myelination of the delay lines peaks at the third week posthatch, and myelinating oligodendrocyte density approaches adult levels by one month, when the head reaches its adult width. Migration of oligodendrocyte progenitors into NL and the subsequent onset of myelination may be restricted by a glial barrier in late embryonic stages and the first posthatch week, since the loss of tenascin-C immunoreactivity in NL is correlated with oligodendrocyte progenitor migration into NL.
Collapse
Affiliation(s)
- Shih-Min Cheng
- Department of Biology, University of Maryland, College Park, MD 20742-4415, USA.
| | | |
Collapse
|
33
|
Origin and progeny of reactive gliosis: A source of multipotent cells in the injured brain. Proc Natl Acad Sci U S A 2008; 105:3581-6. [PMID: 18299565 DOI: 10.1073/pnas.0709002105] [Citation(s) in RCA: 557] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Reactive gliosis is the universal reaction to brain injury, but the precise origin and subsequent fate of the glial cells reacting to injury are unknown. Astrocytes react to injury by hypertrophy and up-regulation of the glial-fibrillary acidic protein (GFAP). Whereas mature astrocytes do not normally divide, a subpopulation of the reactive GFAP(+) cells does so, prompting the question of whether the proliferating GFAP(+) cells arise from endogenous glial progenitors or from mature astrocytes that start to proliferate in response to brain injury. Here we show by genetic fate mapping and cell type-specific viral targeting that quiescent astrocytes start to proliferate after stab wound injury and contribute to the reactive gliosis and proliferating GFAP(+) cells. These proliferating astrocytes remain within their lineage in vivo, while a more favorable environment in vitro revealed their multipotency and capacity for self-renewal. Conversely, progenitors present in the adult mouse cerebral cortex labeled by NG2 or the receptor for the platelet-derived growth factor (PDGFRalpha) did not form neurospheres after (or before) brain injury. Taken together, the first fate-mapping analysis of astrocytes in the adult mouse cerebral cortex shows that some astrocytes acquire stem cell properties after injury and hence may provide a promising cell type to initiate repair after brain injury.
Collapse
|
34
|
von Holst A. Tenascin C in stem cell niches: redundant, permissive or instructive? Cells Tissues Organs 2007; 188:170-7. [PMID: 18160825 DOI: 10.1159/000112848] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The stem cell niche provides the specialized environment that is able to sustain the lifelong maintenance of stem cells in their discrete locations within organs. The niche is usually composed of several different cell types and a specialized extracellular matrix consisting of many different constituents. Additionally, a variety of growth factors are secreted into the extracellular space and contribute to the functional organization of the niche. Here, I will concentrate on the multimodular extracellular matrix glycoprotein tenascin C (Tnc) and discuss it as an exemplary molecule that is present in several stem cell niches. In spite of its intuitively suggestive presence, it has been difficult to provide functional evidence for the importance of Tnc in the context of stem cells. In the nervous system, the careful analysis of Tnc-deficient mice has revealed that the developmental program neural stem cell pass-through is delayed due to changes in growth factor responsiveness. To gain further insight, we have employed the gene trap technology in neural stem cells to identify potential Tnc target genes. This approach has surfaced 2 interesting candidates that may contribute to a better understanding of the signal(s) elicited by Tnc in neural stem/progenitor cells in the niche.
Collapse
Affiliation(s)
- Alexander von Holst
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
35
|
Malatesta P, Appolloni I, Calzolari F. Radial glia and neural stem cells. Cell Tissue Res 2007; 331:165-78. [PMID: 17846796 DOI: 10.1007/s00441-007-0481-8] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Accepted: 07/17/2007] [Indexed: 01/19/2023]
Abstract
During the last decade, the role of radial glia has been radically revisited. Rather than being considered a mere structural component serving to guide newborn neurons towards their final destinations, radial glia is now known to be the main source of neurons in several regions of the central nervous system, notably in the cerebral cortex. Radial glial cells differentiate from neuroepithelial progenitors at the beginning of neurogenesis and share with their ancestors the bipolar shape and the expression of some molecular markers. Radial glia, however, can be distinguished from neuroepithelial progenitors by the expression of astroglial markers. Clonal analyses showed that radial glia is a heterogeneous population, comprising both pluripotent and different lineage-restricted neural progenitors. At late-embryonic and postnatal stages, radial glial cells give rise to the neural stem cells responsible for adult neurogenesis. Embryonic pluripotent radial glia and adult neural stem cells may be clonally linked, thus representing a lineage displaying stem cell features in both the developing and mature central nervous system.
Collapse
Affiliation(s)
- Paolo Malatesta
- Dipartimento di Oncologia, Biologia e Genetica, Università degli Studi di Genova, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | | | | |
Collapse
|
36
|
Zappaterra MD, Lisgo SN, Lindsay S, Gygi SP, Walsh CA, Ballif BA. A Comparative Proteomic Analysis of Human and Rat Embryonic Cerebrospinal Fluid. J Proteome Res 2007; 6:3537-48. [PMID: 17696520 DOI: 10.1021/pr070247w] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
During vertebrate central nervous system development, the apical neuroepithelium is bathed with embryonic Cerebrospinal Fluid (e-CSF) which plays regulatory roles in cortical cell proliferation and maintenance. Here, we report the first proteomic analysis of human e-CSF and compare it to an extensive proteomic analysis of rat e-CSF. As expected, we identified a large collection of protease inhibitors, extracellular matrix proteins, and transport proteins in CSF. However, we also found a surprising suite of signaling and intracellular proteins not predicted by previous proteomic analysis. Some of the intracellular proteins are likely to represent the contents of microvesicles recently described within the CSF (Marzesco, A. M., et al. J. Cell Sci. 2005, 118 (Pt. 13), 2849-2858). Defining the rich composition of e-CSF will enable a greater understanding of its concerted actions during critical stages of brain development.
Collapse
Affiliation(s)
- Mauro D Zappaterra
- Division of Genetics, Children's Hospital Boston, Howard Hughes Medical Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
37
|
Sirko S, von Holst A, Wizenmann A, Götz M, Faissner A. Chondroitin sulfate glycosaminoglycans control proliferation, radial glia cell differentiation and neurogenesis in neural stem/progenitor cells. Development 2007; 134:2727-38. [PMID: 17596283 DOI: 10.1242/dev.02871] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Although the local environment is known to regulate neural stem cell (NSC) maintenance in the central nervous system, little is known about the molecular identity of the signals involved. Chondroitin sulfate proteoglycans (CSPGs) are enriched in the growth environment of NSCs both during development and in the adult NSC niche. In order to gather insight into potential biological roles of CSPGs for NSCs, the enzyme chondroitinase ABC (ChABC) was used to selectively degrade the CSPG glycosaminoglycans. When NSCs from mouse E13 telencephalon were cultivated as neurospheres, treatment with ChABC resulted in diminished cell proliferation and impaired neuronal differentiation, with a converse increase in astrocytes. The intrauterine injection of ChABC into the telencephalic ventricle at midneurogenesis caused a reduction in cell proliferation in the ventricular zone and a diminution of self-renewing radial glia, as revealed by the neurosphere-formation assay, and a reduction in neurogenesis. These observations suggest that CSPGs regulate neural stem/progenitor cell proliferation and intervene in fate decisions between the neuronal and glial lineage.
Collapse
Affiliation(s)
- Swetlana Sirko
- Chair of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Building NDEF 05/339, Universitaetsstrasse 150, D-44780 Bochum, Germany
| | | | | | | | | |
Collapse
|
38
|
von Holst A, Egbers U, Prochiantz A, Faissner A. Neural Stem/Progenitor Cells Express 20 Tenascin C Isoforms That Are Differentially Regulated by Pax6. J Biol Chem 2007; 282:9172-81. [PMID: 17264084 DOI: 10.1074/jbc.m608067200] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tenascin C (Tnc) is an alternatively spliced, multimodular extracellular matrix glycoprotein present in the ventricular zone of the developing brain. Pax6-deficient small eye (sey) mouse mutants show an altered Tnc expression pattern. Here, we investigated the expression of Tnc isoforms in neural stem/progenitor cells and their regulation by the paired-box transcription factor Pax6. Neural stem/progenitor cells cultured as neurospheres strongly expressed Tnc on the protein level. The Tnc isoform expression in neural stem/progenitor cells was analyzed by reverse transcriptase-PCR and dot blot Southern hybridization. In total, 20 different Tnc isoforms were detected in neurospheres derived from embryonic fore-brain cell suspensions. The Tnc isoform containing the fibronectin type III domains A1A4BD is novel and might be neural stem/progenitor cell-specific. Transient overexpression of Pax6 in neurospheres of the medial ganglionic eminence did not alter the total Tnc mRNA expression level but showed a pronounced regulative effect on different Tnc isoforms. The larger Tnc isoforms containing four, five, and six additional alternatively spliced fibronectin type III domains were up-regulated, whereas the small Tnc isoforms without any or with one additional domain were down-regulated. Thus, Pax6 is a homeodomain protein that also modulates the splicing machinery. We conclude that the combinatorial code of Tnc isoform expression in the neural stem/progenitor cell is complex and regulated by Pax6. These findings suggest a functional significance for individual Tnc isoforms in neural stem/progenitor cells.
Collapse
Affiliation(s)
- Alexander von Holst
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, D-44780 Bochum, Germany.
| | | | | | | |
Collapse
|
39
|
Ajioka I, Maeda T, Nakajima K. Identification of ventricular-side-enriched molecules regulated in a stage-dependent manner during cerebral cortical development. Eur J Neurosci 2006; 23:296-308. [PMID: 16420439 DOI: 10.1111/j.1460-9568.2005.04544.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Radial glial cells are the main component of the embryonic cortical ventricular zone (VZ), producing deep-layer excitatory neurons in the early stage and upper-layer excitatory neurons in the late stage of development. Previous studies have suggested that the laminar fate of deep-layer neurons might be determined by early-stage-specific secretory or transmembrane molecules (S/TMs) in the VZ. However, the different properties required to produce the different types of neurons in early-stage and late-stage VZ cells are largely unknown. Herein, we investigated the stage-dependent transcriptional profiles of the ventricular side of the mouse cortex, which was manually dissected at embryonic day (E)12, E14 and E16, and identified 3985 'VZ-enriched' genes, regulated stage-dependently, by GeneChip analysis. These molecules were classified into nine types based on stage-dependent regulation patterns. Prediction programs for the S/TMs revealed 659 'VZ-enriched' S/TMs. In situ hybridization and real-time PCR analysis for several of these molecules showed results consistent with the statistical analysis of the GeneChip experiments. Moreover, we identified 17 cell cycle-related early-stage and 'VZ-enriched' molecules. These molecules included not only those involved in cell cycle progression, but also essential molecules for DNA double-strand break repair, such as Rad51 and Rpa1. These results suggest that the early stage-VZ cells, which produce both deep- and upper-layer neurons, and the late-stage VZ cells, which produce only upper-layer neurons, are intrinsically different. The gene lists presented here will be useful for the investigation of stage-dependent changes in VZ cells and their regulatory mechanisms in the developing cortex.
Collapse
Affiliation(s)
- Itsuki Ajioka
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | |
Collapse
|
40
|
Guntinas-Lichius O, Angelov DN, Morellini F, Lenzen M, Skouras E, Schachner M, Irintchev A. Opposite impacts of tenascin-C and tenascin-R deficiency in mice on the functional outcome of facial nerve repair. Eur J Neurosci 2006; 22:2171-9. [PMID: 16262655 DOI: 10.1111/j.1460-9568.2005.04424.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The glycoproteins tenascin-C (TNC) and tenascin-R (TNR) are extracellular matrix proteins involved in the development, plasticity and repair of the nervous system. Altered expression patterns after nerve lesions in adult animals have suggested that these molecules influence axonal regeneration. To test this hypothesis, we investigated adult mice constitutively deficient in the expression of TNC, TNR or both, using the facial nerve injury paradigm. Quantitative analysis of vibrissal movements prior to nerve transection and repair (facial-facial anastomosis) did not reveal genotype-specific differences, and thus impacts of the mutations on motor function in intact animals. Two months after nerve repair, recovery of vibrissal whisking was poor in wild-type mice, a typical finding after facial-facial anastomosis in rodents. Differential effects of the mutations on whisking were found: recovery of function was worse in TNC-deficient and better in TNR null mice compared with wild-type littermates. In double-knockout animals, vibrissal performance was insufficient, but to a lesser extent compared with TNC null mutant mice. Retrograde labelling of motoneurons in the same animals showed that similar numbers of motoneurons had reinnervated the whisker pads in all experimental groups precluding varying extents of motoneuron death and/or axon regeneration failures as causes for the different outcomes of nerve repair. Our results provide strong evidence that TNC promotes and TNR impedes recovery after nerve lesion. These findings are of particular interest with regard to the scanty knowledge about factors determining success of regeneration in the peripheral nervous system of mammals.
Collapse
|
41
|
Garwood J, Garcion E, Dobbertin A, Heck N, Calco V, ffrench-Constant C, Faissner A. The extracellular matrix glycoprotein Tenascin-C is expressed by oligodendrocyte precursor cells and required for the regulation of maturation rate, survival and responsiveness to platelet-derived growth factor. Eur J Neurosci 2005; 20:2524-40. [PMID: 15548197 DOI: 10.1111/j.1460-9568.2004.03727.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Analysis of Tenascin-C (TN-C) knockout mice revealed novel roles for this extracellular matrix (ECM) protein in regulation of the developmental programme of oligodendrocyte precursor cells (OPCs), their maturation into myelinating oligodendrocytes and sensitivity to growth factors. A major component of the ECM of developing nervous tissue, TN-C was expressed in zones of proliferation, migration and morphogenesis. Examination of TN-C knockout mice showed roles for TN-C in control of OPC proliferation and migration towards zones of myelination [E. Garcion et al. (2001) Development, 128, 2485-2496]. Extending our studies of TN-C effects on OPC development we found that OPCs can endogenously express TN-C protein. This expression covered the whole range of possible TN-C isoforms and could be strongly up-regulated by leukaemia inhibitory factor and ciliary neurotrophic factor, cytokines known to modulate OPC proliferation and survival. Comparative analysis of TN-C knockout OPCs with wild-type OPCs reveals an accelerated rate of maturation in the absence of TN-C, with earlier morphological differentiation and precocious expression of myelin basic protein. TN-C knockout OPCs plated on poly-lysine displayed higher levels of apoptosis than wild-type OPCs and there was also an earlier loss of responsiveness to the protective effects of platelet-derived growth factor (PDGF), indicating that TN-C has anti-apoptotic effects that may be associated with PDGF signalling. The existence of mechanisms to compensate for the absence of TN-C in the knockout is indicated by the development of oligodendrocytes derived from TN-C knockout neurospheres. These were present in equivalent proportions to those found in wild-type neurospheres but displayed enhanced myelin membrane formation.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens/metabolism
- Blotting, Western/methods
- Brain/cytology
- Brain/embryology
- Brain/growth & development
- Brain/metabolism
- Bromodeoxyuridine/metabolism
- Cell Count/methods
- Cell Differentiation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Cerebral Cortex/cytology
- Chondroitin Sulfates/metabolism
- Cytokines/pharmacology
- Embryo, Mammalian
- Gene Expression Regulation, Developmental/drug effects
- Humans
- Immunohistochemistry/methods
- In Situ Hybridization/methods
- In Situ Nick-End Labeling/methods
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Biological
- Myelin Basic Protein/metabolism
- Nerve Tissue Proteins/metabolism
- Oligodendroglia/drug effects
- Oligodendroglia/metabolism
- Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology
- Platelet-Derived Growth Factor/pharmacology
- Protein Tyrosine Phosphatases/metabolism
- Proteoglycans/metabolism
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Receptor-Like Protein Tyrosine Phosphatases, Class 5
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Stem Cells/drug effects
- Stem Cells/metabolism
- Tenascin/genetics
- Tenascin/physiology
- Time Factors
Collapse
Affiliation(s)
- Jeremy Garwood
- LNDR, CNRS 5, rue Blaise Pascal, 67084 Strasbourg Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
42
|
Irintchev A, Rollenhagen A, Troncoso E, Kiss JZ, Schachner M. Structural and functional aberrations in the cerebral cortex of tenascin-C deficient mice. ACTA ACUST UNITED AC 2004; 15:950-62. [PMID: 15537675 DOI: 10.1093/cercor/bhh195] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The extracellular matrix glycoprotein tenascin-C (TNC) has been implicated in neural development and plasticity but many of its functions in vivo remain obscure. Here we addressed the question as to whether the constitutive absence of TNC in mice affects cortical physiology and structure. Defined major cell populations (neurons and inhibitory neuronal subpopulations, astrocytes, oligodendrocytes and microglia) were quantified in the somatosensory and motor cortices of adult TNC deficient (TNC-/-) and wild-type (TNC+/+) mice by immunofluorescence labelling and stereology. In both areas studied we found abnormally high neuronal density, astrogliosis, low density of parvalbumin-positive interneurons and reduced ratios of oligodendrocytes to neurons and of inhibitory to excitatory neurons in the TNC deficient as opposed to the non-deficient animals. Analysis of Golgi-impregnated layer V pyramidal neurons in TNC-/- animals showed aberrant dendrite tortuosity and redistribution of stubby spines within first- to third-order dendritic arbors. Significantly enhanced responses upon whisker stimulation were recorded epicranially over the barrel and the motor cortices of TNC-/- as compared to TNC+/+ animals, and this effect might be associated with the diminished inhibitory circuitry. These results indicate that TNC is essential for normal cortical development and function.
Collapse
Affiliation(s)
- Andrey Irintchev
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, D-20246 Hamburg, Germany
| | | | | | | | | |
Collapse
|
43
|
Garcion E, Halilagic A, Faissner A, ffrench-Constant C. Generation of an environmental niche for neural stem cell development by the extracellular matrix molecule tenascin C. Development 2004; 131:3423-32. [PMID: 15226258 DOI: 10.1242/dev.01202] [Citation(s) in RCA: 227] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Stem cells in the embryonic mammalian CNS are initially responsive to fibroblast growth factor 2 (FGF2). They then undergo a developmental programme in which they acquire epidermal growth factor (EGF) responsiveness, switch from the production of neuronal to glial precursors and become localized in specialized germinal zones such as the subventricular zone (SVZ). Here we show that extracellular matrix molecules act as regulators of this programme. Tenascin C is highly expressed in the SVZ, and transgenic mice lacking tenascin C show delayed acquisition of the EGF receptor. This results from alterations in the response of the stem cells to the growth factors FGF2 and bone morphogenic protein 4 (BMP4), which normally promote and inhibit acquisition of the EGF receptor, respectively. Tenascin C-deficient mice also have altered numbers of CNS stem cells and these stem cells have an increased probability of generating neurones when grown in cell culture. We conclude that tenascin C contributes to the generation of a stem cell 'niche' within the SVZ, acting to orchestrate growth factor signalling so as to accelerate neural stem cell development.
Collapse
Affiliation(s)
- Emmanuel Garcion
- Cambridge Centre for Brain Repair, and Department of Medical Genetics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | | | | | | |
Collapse
|
44
|
Bolz J, Uziel D, Mühlfriedel S, Güllmar A, Peuckert C, Zarbalis K, Wurst W, Torii M, Levitt P. Multiple roles of ephrins during the formation of thalamocortical projections: Maps and more. ACTA ACUST UNITED AC 2004; 59:82-94. [PMID: 15007829 DOI: 10.1002/neu.10346] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The functional architecture of the cerebral cortex is based on intrinsic connections that precisely link neurons from distinct cortical laminae as well as layer-specific afferent and efferent projections. Experimental strategies using in vitro assays originally developed by Friedrich Bonhoeffer have suggested that positional cues confined to individual layers regulate the assembly of local cortical circuits and the formation of thalamocortical projections. One of these wiring molecules is ephrinA5, a ligand for Eph receptor tyrosine kinases. EphrinA5 and Eph receptors exhibit highly dynamic expression patterns in distinct regions of the cortex and thalamus during early and late stages of thalamocortical and cortical circuit formation. In vitro assays suggest that ephrinA5 is a multifunctional wiring molecule for different populations of cortical and thalamic axons. Additionally, the expression patterns of ephrinA5 during cortical development are consistent with this molecule regulating, in alternative ways, specific components of thalamic and cortical connectivity. To test this directly, the organization of thalamocortical projections was examined in mice lacking ephrinA5 gene expression. The anatomical studies in ephrinA5 knockout animals revealed a miswiring of limbic thalamic projections and changes in neocortical circuits that were predicted from the expression pattern and the in vitro analysis of ephrinA5 function.
Collapse
Affiliation(s)
- Jürgen Bolz
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, Erberstrasse 1, 07743 Jena, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Samuel S, Bernstein LR. Adhesion, migration, transcriptional, interferon-inducible, and other signaling molecules newly implicated in cancer susceptibility and resistance of JB6 cells by cDNA microarray analyses. Mol Carcinog 2003; 39:34-60. [PMID: 14694446 DOI: 10.1002/mc.10163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Relative expression levels of 9500 genes were determined by cDNA microarray analyses in mouse skin JB6 cells susceptible (P+) and resistant (P-) to 12-O-tetradecanoyl phorbol-13 acetate (TPA)-induced neoplastic transformation. Seventy-four genes in 6 functional classes were differentially expressed: (I) extracellular matrix (ECM) and basement membrane (BM) proteins (20 genes). P+ cells express higher levels than P- cells of several collagens and proteases, and lower levels of protease inhibitors. Multiple genes encoding adhesion molecules are expressed preferentially in P- cells, including six genes implicated in axon guidance and adhesion. (II) Cytoskeletal proteins (13 genes). These include actin isoforms and regulatory proteins, almost all preferentially expressed in P- cells. (III) Signal transduction proteins (12 genes). Among these are Ras-GTPase activating protein (Ras-GAP), the deleted in oral cancer-1 and SLIT2 tumor suppressors, and connexin 43 (Cx43) gap junctional protein, all expressed preferentially in P- cells. (IV) Interferon-inducible proteins (3 genes). These include interferon-inducible protein (IFI)-16, an Sp1 transcriptional regulator expressed preferentially in P- cells. (V) Other transcription factors (4 genes). Paired related homeobox gene 2 (Prx2)/S8 homeobox, and retinoic acid (RA)-regulated nur77 and cellular retinoic acid-binding protein II (CRABPII) transcription factors are expressed preferentially in P- cells. The RIN-ZF Sp-transcriptional suppressor exhibits preferential P+ expression. (VI) Genes of unknown functions (22 sequences). Numerous mesenchymal markers are expressed in both cell types. Data for multiple genes were confirmed by real-time PCR. Overall, 26 genes were newly implicated in cancer. Detailed analyses of the functions of the genes and their interrelationships provided converging evidence for their possible roles in implementing genetic programs mediating cancer susceptibility and resistance. These results, in conjunction with cell wounding and phalloidin staining data, indicated that concerted genetic programs were implemented that were conducive to cell adhesion and tumor suppression in P- cells and that favored matrix turnover, cell motility, and abrogation of tumor suppression in P+ cells. Such genetic programs may in part be orchestrated by Sp-, RA-, and Hox-transcriptional regulatory pathways implicated in this study.
Collapse
Affiliation(s)
- Shaija Samuel
- Department of Pathology and Laboratory Medicine, Texas A & M University System Health Science Center, College Station, Texas, USA
| | | |
Collapse
|
46
|
Garcion E, Faissner A, ffrench-Constant C. La ténascine-C : une molécule de la matrice extracellulaire impliquée dans le développement du système nerveux central. Med Sci (Paris) 2002. [DOI: 10.1051/medsci/20021810982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
47
|
Chapouton P, Schuurmans C, Guillemot F, Götz M. The transcription factor neurogenin 2 restricts cell migration from the cortex to the striatum. Development 2001; 128:5149-59. [PMID: 11748150 DOI: 10.1242/dev.128.24.5149] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The dorsal and ventral domains of the telencephalon are delineated by a unique boundary structure that restricts the migration of dorsal and ventral cells to a different extent. While many cells invade the dorsal cortex from the ventral ganglionic eminence (GE), hardly any cortical cells cross the boundary into the GE. Several molecules have been implicated in the regulation of ventral to dorsal cell migration, but so far nothing is known about the molecular mechanisms restricting cortical cell migration in vivo. Here we show that in the absence of the transcription factor neurogenin 2, cells from the cortex migrate into the GE in vitro and in vivo as detected in transgenic mice containing a lacZ gene in the neurogenin 2 locus. In contrast, the migration of cells from the GE is not affected. Molecular and cellular analysis of the cortico-striatal boundary revealed that neurogenin 2 regulates the fasciculation of the cortico-striatal boundary which may explain the non cell-autonomous nature of the migration defect as detected by in vitro transplantation. Taken together, these results show that distinct cues located in the cortico-striatal boundary restrict cells in the dorsal and ventral telencephalon.
Collapse
Affiliation(s)
- P Chapouton
- Max-Planck Institute for Neurobiology, Am Klopferspitz 18A, D-82152 Planegg-Martinsried, Germany
| | | | | | | |
Collapse
|
48
|
Keith CH, Wilson MT. Factors controlling axonal and dendritic arbors. INTERNATIONAL REVIEW OF CYTOLOGY 2001; 205:77-147. [PMID: 11336394 DOI: 10.1016/s0074-7696(01)05003-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The sculpting and maintenance of axonal and dendritic arbors is largely under the control of molecules external to the cell. These factors include both substratum-associated and soluble factors that can enhance or inhibit the outgrowth of axons and dendrites. A large number of factors that modulate axonal outgrowth have been identified, and the first stages of the intracellular signaling pathways by which they modify process outgrowth have been characterized. Relatively fewer factors and pathways that affect dendritic outgrowth have been described. The factors that affect axonal arbors form an incompletely overlapping set with those that affect dendritic arbors, allowing selective control of the development and maintenance of these critical aspects of neuronal morphology.
Collapse
Affiliation(s)
- C H Keith
- Department of Cellular Biology. University of Georgia, Athens, 30605, USA
| | | |
Collapse
|
49
|
Identification of a neurite outgrowth-promoting motif within the alternatively spliced region of human tenascin-C. J Neurosci 2001. [PMID: 11549732 DOI: 10.1523/jneurosci.21-18-07215.2001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Our work centers on understanding how the extracellular matrix molecule tenascin-C regulates neuronal growth. We have found that the region of tenascin-C containing only alternately spliced fibronectin type-III repeat D, called fnD, when used by itself, dramatically increases neurite outgrowth in culture. We used overlapping synthetic peptides to localize the neurite outgrowth-promoting site within fnD to a 15 amino acid sequence, called D5. An antibody against D5 blocked promotion of neurite outgrowth by fnD as well as tenascin-C, indicating that this peptide sequence is functional in the context of the native molecule. Further testing of shorter synthetic peptides restricted the neurite outgrowth-promoting site to eight amino acids, VFDNFVLK. Of these, "FD" and "FV" are conserved in tenascin-C sequences derived from all the species available in the GenBank. To investigate the hypothesis that FD and FV are critical for the interaction with neurons, we tested a recombinant fnD protein and synthetic peptides with alterations in FD and/or FV. These molecules did not facilitate process extension, suggesting that the conserved amino acids are required for formation of the active site in fnD. We next investigated whether VFDNFVLK could be used as a reagent to overcome the neurite outgrowth inhibitory properties of chondroitin sulfate proteoglycans, the major inhibitory molecules in the glial scar. The peptide significantly enhanced outgrowth on proteoglycans and was more effective than laminin-1, L1-Fc, or intact tenascin-C, thus demonstrating the potential applicability of tenascin-C regions as therapeutic reagents.
Collapse
|
50
|
Alvarez-Dolado M, Figueroa A, Kozlov S, Sonderegger P, Furley AJ, Muñoz A. Thyroid hormone regulates TAG-1 expression in the developing rat brain. Eur J Neurosci 2001; 14:1209-18. [PMID: 11703450 DOI: 10.1046/j.0953-816x.2001.01745.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
TAG-1 is a member of the immunoglobulin superfamily of cell adhesion molecules thought to play important roles in neuronal differentiation and the establishment of connectivity during brain development. Because these are processes also affected by hypothyroidism, we studied the effects of thyroid hormone deprivation and administration on TAG-1 expression in the developing rat brain. By in situ hybridization, immunohistochemistry and Western blotting we found that TAG-1 RNA and protein levels are upregulated in the hypothyroid brain. From embryonic day 20 to postnatal day (P) 15, elevated TAG-1 RNA was found in several areas including the cerebral cortex, hippocampus and olfactory bulb. In agreement with this, TAG-1 protein was overexpressed in the major fibre tracts arising from these structures, including the corpus callosum, anterior and hippocampal commissures and lateral olfactory tract. A similar overexpression of TAG-1 by hypothyroidism was detected in the cerebellum, but starting only at P15. In all cases, elevation of TAG-1 RNA and protein expression could be reversed by thyroid hormone treatment. These results show that the deregulation of TAG-1 might contribute to the alterations caused by the lack of thyroid hormone during brain development.
Collapse
Affiliation(s)
- M Alvarez-Dolado
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Autónoma de Madrid (UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | | | | | | | | | | |
Collapse
|