1
|
Hutchings C, Nuriel Y, Lazar D, Kohl A, Muir E, Genin O, Cinnamon Y, Benyamini H, Nevo Y, Sela-Donenfeld D. Hindbrain boundaries as niches of neural progenitor and stem cells regulated by the extracellular matrix proteoglycan chondroitin sulphate. Development 2024; 151:dev201934. [PMID: 38251863 PMCID: PMC10911165 DOI: 10.1242/dev.201934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/12/2024] [Indexed: 01/23/2024]
Abstract
The interplay between neural progenitors and stem cells (NPSCs), and their extracellular matrix (ECM) is a crucial regulatory mechanism that determines their behavior. Nonetheless, how the ECM dictates the state of NPSCs remains elusive. The hindbrain is valuable to examine this relationship, as cells in the ventricular surface of hindbrain boundaries (HBs), which arise between any two neighboring rhombomeres, express the NPSC marker Sox2, while being surrounded with the membrane-bound ECM molecule chondroitin sulphate proteoglycan (CSPG), in chick and mouse embryos. CSPG expression was used to isolate HB Sox2+ cells for RNA-sequencing, revealing their distinguished molecular properties as typical NPSCs, which express known and newly identified genes relating to stem cells, cancer, the matrisome and cell cycle. In contrast, the CSPG- non-HB cells, displayed clear neural-differentiation transcriptome. To address whether CSPG is significant for hindbrain development, its expression was manipulated in vivo and in vitro. CSPG manipulations shifted the stem versus differentiation state of HB cells, evident by their behavior and altered gene expression. These results provide further understanding of the uniqueness of hindbrain boundaries as repetitive pools of NPSCs in-between the rapidly growing rhombomeres, which rely on their microenvironment to maintain their undifferentiated state during development.
Collapse
Affiliation(s)
- Carmel Hutchings
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Yarden Nuriel
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Daniel Lazar
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Elizabeth Muir
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 1TN, UK
| | - Olga Genin
- Agricultural Research Organization, Volcani Center, Department of Poultry and Aquaculture Science, Rishon LeTsiyon 7505101, Israel
| | - Yuval Cinnamon
- Agricultural Research Organization, Volcani Center, Department of Poultry and Aquaculture Science, Rishon LeTsiyon 7505101, Israel
| | - Hadar Benyamini
- Info-CORE, Bioinformatics Unit of the I-CORE at the Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yuval Nevo
- Info-CORE, Bioinformatics Unit of the I-CORE at the Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
2
|
Glotzbach K, Faissner A. Substrate-bound and soluble domains of tenascin-C regulate differentiation, proliferation and migration of neural stem and progenitor cells. Front Cell Neurosci 2024; 18:1357499. [PMID: 38425428 PMCID: PMC10902920 DOI: 10.3389/fncel.2024.1357499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction The lack of regenerative capacity of the central nervous system is one of the major challenges nowadays. The knowledge of guidance cues that trigger differentiation, proliferation, and migration of neural stem and progenitor cells is one key element in regenerative medicine. The extracellular matrix protein tenascin-C (Tnc) is a promising candidate to regulate cell fate due to its expression in the developing central nervous system and in the adult neural stem cell niches. Of special interest are the alternatively spliced fibronectin type III (FnIII) domains of Tnc whose combinatorial diversity could theoretically generate up to 64 isoforms in the mouse. A total of 27 isoforms have already been discovered in the developing brain, among others the domain combinations A1D, CD, and A124BCD. Methods In the present study, these domains as well as the combination of the constitutively expressed FnIII domains 7 and 8 (78) were expressed in Chinese hamster ovary cells as pseudo-antibodies fused to the Fc-fragment of a human immunoglobulin G antibody. The fusion proteins were presented to primary mouse neural stem/progenitor cells (NSPCs) grown as neurospheres, either as coated culture substrates or as soluble additives in vitro. The influence of the domains on the differentiation, proliferation and migration of NSPCs was analyzed. Results We observed that the domain combination A124BCD promoted the differentiation of neurons and oligodendrocytes, whereas the domain A1D supported astrocyte differentiation. The constitutively expressed domain 78 had a proliferation and migration stimulating impact. Moreover, most effects were seen only in one of the presentation modes but not in both, suggesting different effects of the Tnc domains in two- and three-dimensional cultures. Discussion This knowledge about the different effect of the Tnc domains might be used to create artificial three-dimensional environments for cell transplantation. Hydrogels spiked with Tnc-domains might represent a promising tool in regenerative medicine.
Collapse
Affiliation(s)
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
3
|
Woo AM, Sontheimer H. Interactions between astrocytes and extracellular matrix structures contribute to neuroinflammation-associated epilepsy pathology. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1198021. [PMID: 39086689 PMCID: PMC11285605 DOI: 10.3389/fmmed.2023.1198021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/31/2023] [Indexed: 08/02/2024]
Abstract
Often considered the "housekeeping" cells of the brain, astrocytes have of late been rising to the forefront of neurodegenerative disorder research. Identified as crucial components of a healthy brain, it is undeniable that when astrocytes are dysfunctional, the entire brain is thrown into disarray. We offer epilepsy as a well-studied neurological disorder in which there is clear evidence of astrocyte contribution to diseases as evidenced across several different disease models, including mouse models of hippocampal sclerosis, trauma associated epilepsy, glioma-associated epilepsy, and beta-1 integrin knockout astrogliosis. In this review we suggest that astrocyte-driven neuroinflammation, which plays a large role in the pathology of epilepsy, is at least partially modulated by interactions with perineuronal nets (PNNs), highly structured formations of the extracellular matrix (ECM). These matrix structures affect synaptic placement, but also intrinsic neuronal properties such as membrane capacitance, as well as ion buffering in their immediate milieu all of which alters neuronal excitability. We propose that the interactions between PNNs and astrocytes contribute to the disease progression of epilepsy vis a vis neuroinflammation. Further investigation and alteration of these interactions to reduce the resultant neuroinflammation may serve as a potential therapeutic target that provides an alternative to the standard anti-seizure medications from which patients are so frequently unable to benefit.
Collapse
Affiliation(s)
- AnnaLin M. Woo
- Neuroscience Graduate Program, Neuroscience Department, University of Virginia, Charlottesville, VA, United States
| | - Harald Sontheimer
- Neuroscience Department, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
4
|
Cadamuro F, Nicotra F, Russo L. 3D printed tissue models: From hydrogels to biomedical applications. J Control Release 2023; 354:726-745. [PMID: 36682728 DOI: 10.1016/j.jconrel.2023.01.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
The development of new advanced constructs resembling structural and functional properties of human organs and tissues requires a deep knowledge of the morphological and biochemical properties of the extracellular matrices (ECM), and the capacity to reproduce them. Manufacturing technologies like 3D printing and bioprinting represent valuable tools for this purpose. This review will describe how morphological and biochemical properties of ECM change in different tissues, organs, healthy and pathological states, and how ECM mimics with the required properties can be generated by 3D printing and bioprinting. The review describes and classifies the polymeric materials of natural and synthetic origin exploited to generate the hydrogels acting as "inks" in the 3D printing process, with particular emphasis on their functionalization allowing crosslinking and conjugation with signaling molecules to develop bio-responsive and bio-instructive ECM mimics.
Collapse
Affiliation(s)
- Francesca Cadamuro
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy
| | - Francesco Nicotra
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy
| | - Laura Russo
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy; CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY Galway, Ireland.
| |
Collapse
|
5
|
Tewari BP, Chaunsali L, Prim CE, Sontheimer H. A glial perspective on the extracellular matrix and perineuronal net remodeling in the central nervous system. Front Cell Neurosci 2022; 16:1022754. [PMID: 36339816 PMCID: PMC9630365 DOI: 10.3389/fncel.2022.1022754] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
A structural scaffold embedding brain cells and vasculature is known as extracellular matrix (ECM). The physical appearance of ECM in the central nervous system (CNS) ranges from a diffused, homogeneous, amorphous, and nearly omnipresent matrix to highly organized distinct morphologies such as basement membranes and perineuronal nets (PNNs). ECM changes its composition and organization during development, adulthood, aging, and in several CNS pathologies. This spatiotemporal dynamic nature of the ECM and PNNs brings a unique versatility to their functions spanning from neurogenesis, cell migration and differentiation, axonal growth, and pathfinding cues, etc., in the developing brain, to stabilizing synapses, neuromodulation, and being an active partner of tetrapartite synapses in the adult brain. The malleability of ECM and PNNs is governed by both intrinsic and extrinsic factors. Glial cells are among the major extrinsic factors that facilitate the remodeling of ECM and PNN, thereby acting as key regulators of diverse functions of ECM and PNN in health and diseases. In this review, we discuss recent advances in our understanding of PNNs and how glial cells are central to ECM and PNN remodeling in normal and pathological states of the CNS.
Collapse
|
6
|
The Extracellular Matrix Proteins Tenascin-C and Tenascin-R Retard Oligodendrocyte Precursor Maturation and Myelin Regeneration in a Cuprizone-Induced Long-Term Demyelination Animal Model. Cells 2022; 11:cells11111773. [PMID: 35681468 PMCID: PMC9179356 DOI: 10.3390/cells11111773] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
Oligodendrocytes are the myelinating cells of the central nervous system. The physiological importance of oligodendrocytes is highlighted by diseases such as multiple sclerosis, in which the myelin sheaths are degraded and the axonal signal transmission is compromised. In a healthy brain, spontaneous remyelination is rare, and newly formed myelin sheaths are thinner and shorter than the former ones. The myelination process requires the migration, proliferation, and differentiation of oligodendrocyte precursor cells (OPCs) and is influenced by proteins of the extracellular matrix (ECM), which consists of a network of glycoproteins and proteoglycans. In particular, the glycoprotein tenascin-C (Tnc) has an inhibitory effect on the differentiation of OPCs and the remyelination efficiency of oligodendrocytes. The structurally similar tenascin-R (Tnr) exerts an inhibitory influence on the formation of myelin membranes in vitro. When Tnc knockout oligodendrocytes were applied to an in vitro myelination assay using artificial fibers, a higher number of sheaths per single cell were obtained compared to the wild-type control. This effect was enhanced by adding brain-derived neurotrophic factor (BDNF) to the culture system. Tnr−/− oligodendrocytes behaved differently in that the number of formed sheaths per single cell was decreased, indicating that Tnr supports the differentiation of OPCs. In order to study the functions of tenascin proteins in vivo Tnc−/− and Tnr−/− mice were exposed to Cuprizone-induced demyelination for a period of 10 weeks. Both Tnc−/− and Tnr−/− mouse knockout lines displayed a significant increase in the regenerating myelin sheath thickness after Cuprizone treatment. Furthermore, in the absence of either tenascin, the number of OPCs was increased. These results suggest that the fine-tuning of myelin regeneration is regulated by the major tenascin proteins of the CNS.
Collapse
|
7
|
Schaberg E, Götz M, Faissner A. The extracellular matrix molecule tenascin-C modulates cell cycle progression and motility of adult neural stem/progenitor cells from the subependymal zone. Cell Mol Life Sci 2022; 79:244. [PMID: 35430697 PMCID: PMC9013340 DOI: 10.1007/s00018-022-04259-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 02/16/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
Abstract
Adult neurogenesis has been described in two canonical regions of the adult central nervous system (CNS) of rodents, the subgranular zone (SGZ) of the hippocampus and the subependymal zone (SEZ) of the lateral ventricles. The stem cell niche of the SEZ provides a privileged environment composed of a specialized extracellular matrix (ECM) that comprises the glycoproteins tenascin-C (Tnc) and laminin-1 (LN1). In the present study, we investigated the function of these ECM glycoproteins in the adult stem cell niche. Adult neural stem/progenitor cells (aNSPCs) of the SEZ were prepared from wild type (Tnc+/+) and Tnc knockout (Tnc−/−) mice and analyzed using molecular and cell biological approaches. A delayed maturation of aNSPCs in Tnc−/− tissue was reflected by a reduced capacity to form neurospheres in response to epidermal growth factor (EGF). To examine a potential influence of the ECM on cell proliferation, aNSPCs of both genotypes were studied by cell tracking using digital video microscopy. aNSPCs were cultivated on three different substrates, namely, poly-d-lysine (PDL) and PDL replenished with either LN1 or Tnc for up to 6 days in vitro. On each of the three substrates aNSPCs displayed lineage trees that could be investigated with regard to cell cycle length. The latter appeared reduced in Tnc−/− aNSPCs on PDL and LN1 substrates, less so on Tnc that seemed to compensate the absence of the ECM compound to some extent. Close inspection of the lineage trees revealed a subpopulation of late dividing aNSPCslate that engaged into cycling after a notable delay. aNSPCslate exhibited a clearly different morphology, with a larger cell body and conspicuous processes. aNSPCslate reiterated the reduction in cell cycle length on all substrates tested, which was not rescued on Tnc substrates. When the migratory activity of aNSPC-derived progeny was determined, Tnc−/− neuroblasts displayed significantly longer migration tracks. This was traced to an increased rate of migration episodes compared to the wild-type cells that rested for longer time periods. We conclude that Tnc intervenes in the proliferation of aNSPCs and modulates the motility of neuroblasts in the niche of the SEZ.
Collapse
Affiliation(s)
- Elena Schaberg
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, LMU, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Biomedical Center, LMU, Planegg-Martinsried, Germany
- Synergy, Excellence Cluster for Systems Neurology, BMC, LMU, Planegg-Martinsried, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
8
|
Dzyubenko E, Manrique-Castano D, Pillath-Eilers M, Vasileiadou P, Reinhard J, Faissner A, Hermann DM. Tenascin-C restricts reactive astrogliosis in the ischemic brain. Matrix Biol 2022; 110:1-15. [DOI: 10.1016/j.matbio.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/21/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
|
9
|
Bauch J, Ort SV, Ulc A, Faissner A. Tenascins Interfere With Remyelination in an Ex Vivo Cerebellar Explant Model of Demyelination. Front Cell Dev Biol 2022; 10:819967. [PMID: 35372366 PMCID: PMC8965512 DOI: 10.3389/fcell.2022.819967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/24/2022] [Indexed: 01/02/2023] Open
Abstract
Oligodendrocytes form myelin membranes and thereby secure the insulation of axons and the rapid conduction of action potentials. Diseases such as multiple sclerosis highlight the importance of this glial cell population for brain function. In the adult brain, efficient remyelination following the damage to oligodendrocytes is compromised. Myelination is characterized by proliferation, migration, and proper integration of oligodendrocyte precursor cells (OPCs). These processes are among others controlled by proteins of the extracellular matrix (ECM). As a prominent representative ECM molecule, tenascin-C (Tnc) exerts an inhibitory effect on the migration and differentiation of OPCs. The structurally similar paralogue tenascin-R (Tnr) is known to promote the differentiation of oligodendrocytes. The model of lysolecithin-induced demyelination of cerebellar slice cultures represents an important tool for the analysis of the remyelination process. Ex vivo cerebellar explant cultures of Tnc−/− and Tnr−/− mouse lines displayed enhanced remyelination by forming thicker myelin membranes upon exposure to lysolecithin. The inhibitory effect of tenascins on remyelination could be confirmed when demyelinated wildtype control cultures were exposed to purified Tnc or Tnr protein. In that approach, the remyelination efficiency decreased in a dose-dependent manner with increasing concentrations of ECM molecules added. In order to examine potential roles in a complex in vivo environment, we successfully established cuprizone-based acute demyelination to analyze the remyelination behavior after cuprizone withdrawal in SV129, Tnc−/−, and Tnr−/− mice. In addition, we documented by immunohistochemistry in the cuprizone model the expression of chondroitin sulfate proteoglycans that are inhibitory for the differentiation of OPCs. In conclusion, inhibitory properties of Tnc and Tnr for myelin membrane formation could be demonstrated by using an ex vivo approach.
Collapse
|
10
|
Regulating Endogenous Neural Stem Cell Activation to Promote Spinal Cord Injury Repair. Cells 2022; 11:cells11050846. [PMID: 35269466 PMCID: PMC8909806 DOI: 10.3390/cells11050846] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) affects millions of individuals worldwide. Currently, there is no cure, and treatment options to promote neural recovery are limited. An innovative approach to improve outcomes following SCI involves the recruitment of endogenous populations of neural stem cells (NSCs). NSCs can be isolated from the neuroaxis of the central nervous system (CNS), with brain and spinal cord populations sharing common characteristics (as well as regionally distinct phenotypes). Within the spinal cord, a number of NSC sub-populations have been identified which display unique protein expression profiles and proliferation kinetics. Collectively, the potential for NSCs to impact regenerative medicine strategies hinges on their cardinal properties, including self-renewal and multipotency (the ability to generate de novo neurons, astrocytes, and oligodendrocytes). Accordingly, endogenous NSCs could be harnessed to replace lost cells and promote structural repair following SCI. While studies exploring the efficacy of this approach continue to suggest its potential, many questions remain including those related to heterogeneity within the NSC pool, the interaction of NSCs with their environment, and the identification of factors that can enhance their response. We discuss the current state of knowledge regarding populations of endogenous spinal cord NSCs, their niche, and the factors that regulate their behavior. In an attempt to move towards the goal of enhancing neural repair, we highlight approaches that promote NSC activation following injury including the modulation of the microenvironment and parenchymal cells, pharmaceuticals, and applied electrical stimulation.
Collapse
|
11
|
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: 0.5] [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
|
12
|
Schaberg E, Theocharidis U, May M, Lessmann K, Schroeder T, Faissner A. Sulfation of Glycosaminoglycans Modulates the Cell Cycle of Embryonic Mouse Spinal Cord Neural Stem Cells. Front Cell Dev Biol 2021; 9:643060. [PMID: 34169071 PMCID: PMC8217649 DOI: 10.3389/fcell.2021.643060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
In the developing spinal cord neural stem and progenitor cells (NSPCs) secrete and are surrounded by extracellular matrix (ECM) molecules that influence their lineage decisions. The chondroitin sulfate proteoglycan (CSPG) DSD-1-PG is an isoform of receptor protein tyrosine phosphatase-beta/zeta (RPTPβ/ζ), a trans-membrane receptor expressed by NSPCs. The chondroitin sulfate glycosaminoglycan chains are sulfated at distinct positions by sulfotransferases, thereby generating the distinct DSD-1-epitope that is recognized by the monoclonal antibody (mAb) 473HD. We detected the epitope, the critical enzymes and RPTPβ/ζ in the developing spinal cord. To obtain insight into potential biological functions, we exposed spinal cord NSPCs to sodium chlorate. The reagent suppresses the sulfation of glycosaminoglycans, thereby erasing any sulfation code expressed by the glycosaminoglycan polymers. When NSPCs were treated with chlorate and cultivated in the presence of FGF2, their proliferation rate was clearly reduced, while NSPCs exposed to EGF were less affected. Time-lapse video microscopy and subsequent single-cell tracking revealed that pedigrees of NSPCs cultivated with FGF2 were strongly disrupted when sulfation was suppressed. Furthermore, the NSPCs displayed a protracted cell cycle length. We conclude that the inhibition of sulfation with sodium chlorate interferes with the FGF2-dependent cell cycle progression in spinal cord NSPCs.
Collapse
Affiliation(s)
- Elena Schaberg
- Department for Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Ursula Theocharidis
- Department for Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Marcus May
- Department for Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Katrin Lessmann
- Department for Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zürich, Zurich, Switzerland
| | - Andreas Faissner
- Department for Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
13
|
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: 4.8] [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
|
14
|
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: 1.3] [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
|
15
|
Okada T, Suzuki H. The Role of Tenascin-C in Tissue Injury and Repair After Stroke. Front Immunol 2021; 11:607587. [PMID: 33552066 PMCID: PMC7859104 DOI: 10.3389/fimmu.2020.607587] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/04/2020] [Indexed: 12/16/2022] Open
Abstract
Stroke is still one of the most common causes for mortality and morbidity worldwide. Following acute stroke onset, biochemical and cellular changes induce further brain injury such as neuroinflammation, cell death, and blood-brain barrier disruption. Matricellular proteins are non-structural proteins induced by many stimuli and tissue damage including stroke induction, while its levels are generally low in a normal physiological condition in adult tissues. Currently, a matricellular protein tenascin-C (TNC) is considered to be an important inducer to promote neuroinflammatory cascades and the resultant pathology in stroke. TNC is upregulated in cerebral arteries and brain tissues including astrocytes, neurons, and brain capillary endothelial cells following subarachnoid hemorrhage (SAH). TNC may be involved in blood-brain barrier disruption, neuronal apoptosis, and cerebral vasospasm via the activation of mitogen-activated protein kinases and nuclear factor-kappa B following SAH. In addition, post-SAH TNC levels in cerebrospinal fluid predicted the development of delayed cerebral ischemia and angiographic vasospasm in clinical settings. On the other hand, TNC is reported to promote fibrosis and exert repair effects for an experimental aneurysm via macrophages-induced migration and proliferation of smooth muscle cells. The authors review TNC-induced inflammatory signal cascades and the relationships with other matricellular proteins in stroke-related pathology.
Collapse
Affiliation(s)
- Takeshi Okada
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Neurosurgery, Kuwana City Medical Center, Kuwana, Japan
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
16
|
Tenascin-C Function in Glioma: Immunomodulation and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:149-172. [PMID: 32845507 DOI: 10.1007/978-3-030-48457-6_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
First identified in the 1980s, tenascin-C (TNC) is a multi-domain extracellular matrix glycoprotein abundantly expressed during the development of multicellular organisms. TNC level is undetectable in most adult tissues but rapidly and transiently induced by a handful of pro-inflammatory cytokines in a variety of pathological conditions including infection, inflammation, fibrosis, and wound healing. Persistent TNC expression is associated with chronic inflammation and many malignancies, including glioma. By interacting with its receptor integrin and a myriad of other binding partners, TNC elicits context- and cell type-dependent function to regulate cell adhesion, migration, proliferation, and angiogenesis. TNC operates as an endogenous activator of toll-like receptor 4 and promotes inflammatory response by inducing the expression of multiple pro-inflammatory factors in innate immune cells such as microglia and macrophages. In addition, TNC drives macrophage differentiation and polarization predominantly towards an M1-like phenotype. In contrast, TNC shows immunosuppressive function in T cells. In glioma, TNC is expressed by tumor cells and stromal cells; high expression of TNC is correlated with tumor progression and poor prognosis. Besides promoting glioma invasion and angiogenesis, TNC has been found to affect the morphology and function of tumor-associated microglia/macrophages in glioma. Clinically, TNC can serve as a biomarker for tumor progression; and TNC antibodies have been utilized as an adjuvant agent to deliver anti-tumor drugs to target glioma. A better mechanistic understanding of how TNC impacts innate and adaptive immunity during tumorigenesis and tumor progression will open new therapeutic avenues to treat brain tumors and other malignancies.
Collapse
|
17
|
Zabegalov KN, Wang D, Yang L, Wang J, Hu G, Serikuly N, Alpyshov ET, Khatsko SL, Zhdanov A, Demin KA, Galstyan DS, Volgin AD, de Abreu MS, Strekalova T, Song C, Amstislavskaya TG, Sysoev Y, Musienko PE, Kalueff AV. Decoding the role of zebrafish neuroglia in CNS disease modeling. Brain Res Bull 2020; 166:44-53. [PMID: 33027679 DOI: 10.1016/j.brainresbull.2020.09.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
Abstract
Neuroglia, including microglia and astrocytes, is a critical component of the central nervous system (CNS) that interacts with neurons to modulate brain activity, development, metabolism and signaling pathways. Thus, a better understanding of the role of neuroglia in the brain is critical. Complementing clinical and rodent data, the zebrafish (Danio rerio) is rapidly becoming an important model organism to probe the role of neuroglia in brain disorders. With high genetic and physiological similarity to humans and rodents, zebrafish possess some common (shared), as well as some specific molecular biomarkers and features of neuroglia development and functioning. Studying these common and zebrafish-specific aspects of neuroglia may generate important insights into key brain mechanisms, including neurodevelopmental, neurodegenerative, neuroregenerative and neurological processes. Here, we discuss the biology of neuroglia in humans, rodents and fish, its role in various CNS functions, and further directions of translational research into the role of neuroglia in CNS disorders using zebrafish models.
Collapse
Affiliation(s)
- Konstantin N Zabegalov
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia
| | - Dongmei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - LongEn Yang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Jingtao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Guojun Hu
- School of Pharmacy, Southwest University, Chongqing, China
| | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | | | | | | | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia.
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Division of Molecular Psychiatry, Centre of Mental Health, University of Würzburg, Würzburg, Germany
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Tamara G Amstislavskaya
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Zelman Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Yury Sysoev
- Laboratory of Neuroprosthetics, Institute of Translational Biomedicine, Petersburg State University, St. Petersburg, Russia; Department of Pharmacology and Clinical Pharmacology, St. Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| | - Pavel E Musienko
- Laboratory of Neuroprosthetics, Institute of Translational Biomedicine, Petersburg State University, St. Petersburg, Russia; Institute of Phthisiopulmonology, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
18
|
Minta K, Portelius E, Janelidze S, Hansson O, Zetterberg H, Blennow K, Andreasson U. Cerebrospinal Fluid Concentrations of Extracellular Matrix Proteins in Alzheimer's Disease. J Alzheimers Dis 2020; 69:1213-1220. [PMID: 31156172 DOI: 10.3233/jad-190187] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Brevican, neurocan, tenascin-C, and tenascin-R are extracellular matrix (ECM) proteins that are mainly expressed in the brain. They play important roles in proliferation and migration of neurons and other cell types in the brain. These ECM proteins may also be involved in various pathologies, including reactive gliosis. OBJECTIVE The aim of the study was to investigate if ECM protein concentrations in cerebrospinal fluid (CSF) are linked to the neurodegenerative process in Alzheimer's disease (AD). METHODS Lumbar CSF samples from a non-AD control group (n = 50) and a clinically diagnosed AD group (n = 42), matched for age and gender, were analyzed using commercially available ELISAs detecting ECM proteins. Mann-Whitney U test was used to examine group differences, while Spearman's rho test was used for correlations. RESULTS Brevican, neurocan, tenascin-R, and tenascin-C concentrations in AD patients did not differ compared to healthy controls or when the groups were dichotomized based on the Aβ42/40 cut-off. CSF tenascin-C and tenascin-R concentrations were significantly higher in women than in men in the AD group (p = 0.02). CONCLUSION ECM proteins do not reflect AD-pathology in CSF. CSF tenascin-C and tenascin-R upregulation in women possibly reveal sexual dimorphism in the central nervous system immunity during AD.
Collapse
Affiliation(s)
- Karolina Minta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Shorena Janelidze
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Sweden
| | - Oskar Hansson
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Sweden.,Memory Clinic, Skåne University Hospital, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
19
|
Ásgrímsdóttir ES, Arenas E. Midbrain Dopaminergic Neuron Development at the Single Cell Level: In vivo and in Stem Cells. Front Cell Dev Biol 2020; 8:463. [PMID: 32733875 PMCID: PMC7357704 DOI: 10.3389/fcell.2020.00463] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that predominantly affects dopaminergic (DA) neurons of the substantia nigra. Current treatment options for PD are symptomatic and typically involve the replacement of DA neurotransmission by DA drugs, which relieve the patients of some of their motor symptoms. However, by the time of diagnosis, patients have already lost about 70% of their substantia nigra DA neurons and these drugs offer only temporary relief. Therefore, cell replacement therapy has garnered much interest as a potential treatment option for PD. Early studies using human fetal tissue for transplantation in PD patients provided proof of principle for cell replacement therapy, but they also highlighted the ethical and practical difficulties associated with using human fetal tissue as a cell source. In recent years, advancements in stem cell research have made human pluripotent stem cells (hPSCs) an attractive source of material for cell replacement therapy. Studies on how DA neurons are specified and differentiated in the developing mouse midbrain have allowed us to recapitulate many of the positional and temporal cues needed to generate DA neurons in vitro. However, little is known about the developmental programs that govern human DA neuron development. With the advent of single-cell RNA sequencing (scRNA-seq) and bioinformatics, it has become possible to analyze precious human samples with unprecedented detail and extract valuable high-quality information from large data sets. This technology has allowed the systematic classification of cell types present in the human developing midbrain along with their gene expression patterns. By studying human development in such an unbiased manner, we can begin to elucidate human DA neuron development and determine how much it differs from our knowledge of the rodent brain. Importantly, this molecular description of the function of human cells has become and will increasingly be a reference to define, evaluate, and engineer cell types for PD cell replacement therapy and disease modeling.
Collapse
Affiliation(s)
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Haage V, Elmadany N, Roll L, Faissner A, Gutmann DH, Semtner M, Kettenmann H. Tenascin C regulates multiple microglial functions involving TLR4 signaling and HDAC1. Brain Behav Immun 2019; 81:470-483. [PMID: 31271872 DOI: 10.1016/j.bbi.2019.06.047] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 01/15/2023] Open
Abstract
Tenascin C (Tnc) is an extracellular matrix glycoprotein, expressed in the CNS during development, as well as in the setting of inflammation, fibrosis and cancer, which operates as an activator of Toll-like receptor 4 (TLR4). Although TLR4 is highly expressed in microglia, the effect of Tnc on microglia has not been elucidated to date. Herein, we demonstrate that Tnc regulates microglial phagocytic activity at an early postnatal age (P4), and that this process is partially dependent on microglial TLR4 expression. We further show that Tnc regulates proinflammatory cytokine/chemokine production, chemotaxis and phagocytosis in primary microglia in a TLR4-dependent fashion. Moreover, Tnc induces histone-deacetylase 1 (HDAC1) expression in microglia, such that HDAC1 inhibition by MS-275 decreases Tnc-induced microglial IL-6 and TNF-α production. Finally, Tnc-/- cortical microglia have reduced HDAC1 expression levels at P4. Taken together, these findings establish Tnc as a regulator of microglia function during early postnatal development.
Collapse
Affiliation(s)
- Verena Haage
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Nirmeen Elmadany
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Lars Roll
- Zellmorphologie und Molekulare Neurobiologie, Fakultät für Biologie und Biotechnologie, Ruhr-Universität Bochum, Bochum, Nordrhein-Wastfalen 44801, Germany
| | - Andreas Faissner
- Zellmorphologie und Molekulare Neurobiologie, Fakultät für Biologie und Biotechnologie, Ruhr-Universität Bochum, Bochum, Nordrhein-Wastfalen 44801, Germany
| | - David H Gutmann
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.
| |
Collapse
|
21
|
López JM, Morona R, Moreno N, Lozano D, Jiménez S, González A. Pax6 expression highlights regional organization in the adult brain of lungfishes, the closest living relatives of land vertebrates. J Comp Neurol 2019; 528:135-159. [PMID: 31299095 DOI: 10.1002/cne.24744] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/27/2019] [Accepted: 07/05/2019] [Indexed: 12/15/2022]
Abstract
The Pax6 gene encodes a regulatory transcription factor that is key in brain development. The molecular structure of Pax6, the roles it plays and its patterns of expression in the brain have been highly conserved during vertebrate evolution. As neurodevelopment proceeds, the Pax6 expression changes from the mitotic germinal zone in the ventricular zone to become distributed in cell groups in the adult brain. Studies in various vertebrates, from fish to mammals, found that the Pax6 expression is maintained in adults in most regions that express it during development. Specifically, in amphibians, Pax6 is widely expressed in the adult brain and its distribution pattern serves to highlight regional organization of the brain. In the present study, we analyzed the detailed distribution of Pax6 cells in the adult central nervous system of lungfishes, the closest living relatives of all tetrapods. Immunohistochemistry performed using double labeling techniques with several neuronal markers of known distribution patterns served to evaluate the actual location of Pax6 cells. Our results show that the Pax6 expression is maintained in the adult brain of lungfishes, in distinct regions of the telencephalon (pallium and subpallium), diencephalon, mesencephalon, hindbrain, spinal cord, and retina. The pattern of Pax6 expression is largely shared with amphibians and helps to understand the primitive condition that would have characterized the common ancestors to all sarcopterygians (lobe-finned fishes and tetrapods), in which Pax6 would be needed to maintain specific entities of subpopulations of neurons.
Collapse
Affiliation(s)
- Jesús M López
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Ruth Morona
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Nerea Moreno
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Daniel Lozano
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Sara Jiménez
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Agustín González
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| |
Collapse
|
22
|
McNitt DH, Choi SJ, Allen JL, Hames RA, Weed SA, Van De Water L, Berisio R, Lukomski S. Adaptation of the group A Streptococcus adhesin Scl1 to bind fibronectin type III repeats within wound-associated extracellular matrix: implications for cancer therapy. Mol Microbiol 2019; 112:800-819. [PMID: 31145503 PMCID: PMC6736723 DOI: 10.1111/mmi.14317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The human‐adapted pathogen group A Streptococcus (GAS) utilizes wounds as portals of entry into host tissue, wherein surface adhesins interact with the extracellular matrix, enabling bacterial colonization. The streptococcal collagen‐like protein 1 (Scl1) is a major adhesin of GAS that selectively binds to two fibronectin type III (FnIII) repeats within cellular fibronectin, specifically the alternatively spliced extra domains A and B, and the FnIII repeats within tenascin‐C. Binding to FnIII repeats was mediated through conserved structural determinants present within the Scl1 globular domain and facilitated GAS adherence and biofilm formation. Isoforms of cellular fibronectin that contain extra domains A and B, as well as tenascin‐C, are present for several days in the wound extracellular matrix. Scl1‐FnIII binding is therefore an example of GAS adaptation to the host's wound environment. Similarly, cellular fibronectin isoforms and tenascin‐C are present in the tumor microenvironment. Consistent with this, FnIII repeats mediate GAS attachment to and enhancement of biofilm formation on matrices deposited by cancer‐associated fibroblasts and osteosarcoma cells. These data collectively support the premise for utilization of the Scl1‐FnIII interaction as a novel method of anti‐neoplastic targeting in the tumor microenvironment.
Collapse
Affiliation(s)
- Dudley H McNitt
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Soo Jeon Choi
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jessica L Allen
- Department of Biochemistry, Program in Cancer Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - River A Hames
- Department of Biochemistry, Program in Cancer Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Scott A Weed
- Department of Biochemistry, Program in Cancer Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Livingston Van De Water
- Departments of Surgery and Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, USA
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Slawomir Lukomski
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
23
|
Roll L, Faissner A. Tenascins in CNS lesions. Semin Cell Dev Biol 2019; 89:118-124. [DOI: 10.1016/j.semcdb.2018.09.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/03/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023]
|
24
|
Ohayon D, Escalas N, Cochard P, Glise B, Danesin C, Soula C. Sulfatase 2 promotes generation of a spinal cord astrocyte subtype that stands out through the expression of Olig2. Glia 2019; 67:1478-1495. [PMID: 30980466 PMCID: PMC6617735 DOI: 10.1002/glia.23621] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/25/2019] [Accepted: 03/29/2019] [Indexed: 12/12/2022]
Abstract
Generation of glial cell diversity in the developing spinal cord is known to depend on spatio-temporal patterning programs. In particular, expression of the transcription factor Olig2 in neural progenitors of the pMN domain is recognized as critical to their fate choice decision to form oligodendrocyte precursor cells (OPCs) instead of astrocyte precursors (APs). However, generating some confusion, lineage-tracing studies of Olig2 progenitors in the spinal cord provided evidence that these progenitors also generate some astrocytes. Here, we addressed the role of the heparan sulfate-editing enzyme Sulf2 in the control of gliogenesis and found an unanticipated function for this enzyme. At initiation of gliogenesis in mouse, Sulf2 is expressed in ventral neural progenitors of the embryonic spinal cord, including in Olig2-expressing cells of the pMN domain. We found that sulf2 deletion, while not affecting OPC production, impairs generation of a previously unknown Olig2-expressing pMN-derived cell subtype that, in contrast to OPCs, does not upregulate Sox10, PDGFRα or Olig1. Instead, these cells activate expression of AP identity genes, including aldh1L1 and fgfr3 and, of note, retain Olig2 expression as they populate the spinal parenchyma at embryonic stages but also as they differentiate into mature astrocytes at postnatal stages. Thus, our study, by revealing the existence of Olig2-expressing APs that segregate early from pMN cells under the influence of Sulf2, supports the existence of a common source of APs and OPCs in the ventral spinal cord and highlights divergent regulatory mechanism for the development of pMN-derived OPCs and APs.
Collapse
Affiliation(s)
- David Ohayon
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, Toulouse, France
| | - Nathalie Escalas
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, Toulouse, France
| | - Philippe Cochard
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, Toulouse, France
| | - Bruno Glise
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, Toulouse, France
| | - Cathy Danesin
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, Toulouse, France
| | - Cathy Soula
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, Toulouse, France
| |
Collapse
|
25
|
Wilems T, Vardhan S, Wu S, Sakiyama-Elbert S. The influence of microenvironment and extracellular matrix molecules in driving neural stem cell fate within biomaterials. Brain Res Bull 2019; 148:25-33. [PMID: 30898579 DOI: 10.1016/j.brainresbull.2019.03.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/07/2019] [Accepted: 03/12/2019] [Indexed: 12/17/2022]
Abstract
Transplantation of stem cells is a promising potential therapy for central nervous system disease and injury. The capacity for self-renewal, proliferation of progenitor cells, and multi-lineage potential underscores the need for controlling stem cell fate. Furthermore, transplantation within a hostile environment can lead to significant cell death and limited therapeutic potential. Tissue-engineered materials have been developed to both regulate stem cell fate, increase transplanted cell viability, and improve therapeutic outcomes. Traditionally, regulation of stem cell differentiation has been driven through soluble signals, such as growth factors. While these signals are important, insoluble factors from the local microenvironment or extracellular matrix (ECM) molecules also contribute to stem cell activity and fate. Understanding the microenvironment factors that influence stem cell fate, such as mechanical properties, topography, and presentation of specific ECM ligands, is necessary for designing improved biomaterials. Here we review some of the microenvironment factors that regulate stem cell fate and how they can be incorporated into biomaterials as part of potential CNS therapies.
Collapse
Affiliation(s)
- Thomas Wilems
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Sangamithra Vardhan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Siliang Wu
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Shelly Sakiyama-Elbert
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, USA.
| |
Collapse
|
26
|
Mirzaei R, Sarkar S, Dzikowski L, Rawji KS, Khan L, Faissner A, Bose P, Yong VW. Brain tumor-initiating cells export tenascin-C associated with exosomes to suppress T cell activity. Oncoimmunology 2018; 7:e1478647. [PMID: 30288344 DOI: 10.1080/2162402x.2018.1478647] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/01/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022] Open
Abstract
The dismal prognosis of glioblastoma is attributed in part to the existence of stem-like brain tumor-initiating cells (BTICs) that are highly radio- and chemo-resistant. New approaches such as therapies that reprogram compromised immune cells against BTICs are needed. Effective immunotherapies in glioblastoma, however, remain elusive unless the mechanisms of immunosuppression by the tumor are better understood. Here, we describe that while the conditioned media of activated T lymphocytes reduce the growth capacity of BTICs, this growth suppression was abrogated in live co-culture of BTICs with T cells. We present evidence that BTICs produce the extracellular matrix protein tenascin-C (TNC) to inhibit T cell activity in live co-culture. In human glioblastoma brain specimens, TNC was widely deposited in the vicinity of T cells. Mechanistically, TNC inhibited T cell proliferation through interaction with α5β1 and αvβ6 integrins on T lymphocytes associated with reduced mTOR signaling. Strikingly, TNC was exported out of BTICs associated with exosomes, and TNC-depleted exosomes suppressed T cell responses to a significantly lesser extent than control. Finally, we found that circulating exosomes from glioblastoma patients contained more TNC and T cell-suppressive activity than those from control individuals. Taken together, our study establishes a novel immunosuppressive role for TNC associated with BTIC-secreted exosomes to affect local and distal T lymphocyte immunity.
Collapse
Affiliation(s)
- Reza Mirzaei
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Susobhan Sarkar
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Lauren Dzikowski
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Khalil S Rawji
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Lubaba Khan
- Departments of Biochemistry and Molecular Biology, Oncology and Surgery, Ohlson Research Initiative and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Pinaki Bose
- Departments of Biochemistry and Molecular Biology, Oncology and Surgery, Ohlson Research Initiative and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
27
|
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: 1.9] [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
|
28
|
Simitzi C, Karali K, Ranella A, Stratakis E. Controlling the Outgrowth and Functions of Neural Stem Cells: The Effect of Surface Topography. Chemphyschem 2018; 19:1143-1163. [DOI: 10.1002/cphc.201701175] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/19/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Chara Simitzi
- Institute of Electronic Structure and Laser (IESL); Foundation for Research and Technology-Hellas (FORTH); Heraklion 71003 Greece
| | - Kanelina Karali
- Institute of Electronic Structure and Laser (IESL); Foundation for Research and Technology-Hellas (FORTH); Heraklion 71003 Greece
| | - Anthi Ranella
- Institute of Electronic Structure and Laser (IESL); Foundation for Research and Technology-Hellas (FORTH); Heraklion 71003 Greece
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser (IESL); Foundation for Research and Technology-Hellas (FORTH); Heraklion 71003 Greece
| |
Collapse
|
29
|
Su W, Matsumoto S, Sorg B, Sherman LS. Distinct roles for hyaluronan in neural stem cell niches and perineuronal nets. Matrix Biol 2018; 78-79:272-283. [PMID: 29408010 DOI: 10.1016/j.matbio.2018.01.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 12/15/2022]
Abstract
Adult neurogenesis in mammals is a tightly regulated process where neural stem cells (NSCs), especially in the subgranular zone (SGZ) of the hippocampal dentate gyrus, proliferate and differentiate into new neurons that form new circuits or integrate into old circuits involved in episodic memory, pattern discrimination, and emotional responses. Recent evidence suggests that changes in the hyaluronan (HA)-based extracellular matrix of the SGZ may regulate neurogenesis by controlling NSC proliferation and early steps in neuronal differentiation. These studies raise the intriguing possibility that perturbations in this matrix, including HA accumulation with aging, could impact adult neurogenesis and cognitive functions, and that alterations to this matrix could be beneficial following insults to the central nervous system that impact hippocampal functions.
Collapse
Affiliation(s)
- Weiping Su
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Steven Matsumoto
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA; Integrative Biosciences Department, School of Dentistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Barbara Sorg
- Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA 98686, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
30
|
Ohayon D, Garcès A, Joly W, Soukkarieh C, Takagi T, Sabourin JC, Agius E, Darling DS, De Santa Barbara P, Higashi Y, Stolt CC, Hugnot JP, Richardson WD, Carroll P, Pattyn A. Onset of Spinal Cord Astrocyte Precursor Emigration from the Ventricular Zone Involves the Zeb1 Transcription Factor. Cell Rep 2017; 17:1473-1481. [PMID: 27806288 DOI: 10.1016/j.celrep.2016.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 06/17/2016] [Accepted: 10/04/2016] [Indexed: 01/01/2023] Open
Abstract
During spinal cord development, astrocyte precursors arise from neuroepithelial progenitors, delaminate from the ventricular zone, and migrate toward their final locations where they differentiate. Although the mechanisms underlying their early specification and late differentiation are being deciphered, less is known about the temporal control of their migration. Here, we show that the epithelial-mesenchymal transition regulator Zeb1 is expressed in glial precursors and report that loss of Zeb1 function specifically delays the onset of astrocyte precursor delamination from the ventricular zone, correlating with transient deregulation of the adhesion protein Cadherin-1. Consequently, astrocyte precursor invasion into the Zeb1-/- mutant white matter is delayed, and induction of their differentiation is postponed. These findings illustrate how fine regulation of adhesive properties influences the onset of neural precursor migration and further support the notion that duration of exposure of migrating astrocyte precursors to environmental cues and/or their correct positioning influence the timing of their differentiation.
Collapse
Affiliation(s)
- David Ohayon
- INSERM U1051, Institut des Neurosciences de Montpellier, 34091 Montpellier, France; Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), CBD-UMR5547, 31062 Toulouse, France; Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Alain Garcès
- INSERM U1051, Institut des Neurosciences de Montpellier, 34091 Montpellier, France
| | - Willy Joly
- INSERM U1051, Institut des Neurosciences de Montpellier, 34091 Montpellier, France
| | - Chadi Soukkarieh
- INSERM U1051, Institut des Neurosciences de Montpellier, 34091 Montpellier, France; Department of Animal Biology, Faculty of Sciences, Damascus University, Damascus, Syria
| | - Tsuyoshi Takagi
- Department of Perinatology, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan
| | | | - Eric Agius
- Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), CBD-UMR5547, 31062 Toulouse, France
| | - Douglas S Darling
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY 40202, USA
| | - Pascal De Santa Barbara
- PhyMedExp, INSERM U1046, CNRS UMR 9214, University of Montpellier, 34295 Montpellier, France
| | - Yujiro Higashi
- Department of Perinatology, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan
| | - Claus C Stolt
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jean-Philippe Hugnot
- INSERM U1051, Institut des Neurosciences de Montpellier, 34091 Montpellier, France
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Patrick Carroll
- INSERM U1051, Institut des Neurosciences de Montpellier, 34091 Montpellier, France
| | - Alexandre Pattyn
- INSERM U1051, Institut des Neurosciences de Montpellier, 34091 Montpellier, France.
| |
Collapse
|
31
|
Faissner A, Roll L, Theocharidis U. Tenascin-C in the matrisome of neural stem and progenitor cells. Mol Cell Neurosci 2017; 81:22-31. [DOI: 10.1016/j.mcn.2016.11.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 01/16/2023] Open
|
32
|
Kandasamy M, Roll L, Langenstroth D, Brüstle O, Faissner A. Glycoconjugates reveal diversity of human neural stem cells (hNSCs) derived from human induced pluripotent stem cells (hiPSCs). Cell Tissue Res 2017; 368:531-549. [DOI: 10.1007/s00441-017-2594-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 02/23/2017] [Indexed: 12/20/2022]
|
33
|
Fibrin hydrogels induce mixed dorsal/ventral spinal neuron identities during differentiation of human induced pluripotent stem cells. Acta Biomater 2017; 51:237-245. [PMID: 28088670 DOI: 10.1016/j.actbio.2017.01.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 12/21/2022]
Abstract
We hypothesized that generating spinal motor neurons (sMNs) from human induced pluripotent stem cell (hiPSC)-derived neural aggregates (NAs) using a chemically-defined differentiation protocol would be more effective inside of 3D fibrin hydrogels compared to 2D poly-L-ornithine(PLO)/laminin-coated tissue culture plastic surfaces. We performed targeted RNA-Seq using next generation sequencing to determine the substrate-specific differences in gene expression that regulate cell phenotype. Cells cultured on both substrates expressed sMN genes CHAT and MNX1, though persistent WNT signaling contributed to a higher expression of genes associated with interneurons in NAs cultured in 3D fibrin scaffolds. Cells in fibrin also expressed lower levels of astrocyte progenitor genes and higher levels of the neuronal-specific gene TUBB3, suggesting a purer population of neurons compared to 2D cultures. STATEMENT OF SIGNIFICANCE Fibrin scaffolds can support the neuronal differentiation of pluripotent stem cells. This study provides insight into how fibrin hydrogels affect neuronal induction by analyzing of the signaling pathways activated during the differentiation process. These insights can then be used to tailor the properties of these hydrogels to optimize the generation of sMNs for regenerative medicine applications.
Collapse
|
34
|
La Manno G, Gyllborg D, Codeluppi S, Nishimura K, Salto C, Zeisel A, Borm LE, Stott SRW, Toledo EM, Villaescusa JC, Lönnerberg P, Ryge J, Barker RA, Arenas E, Linnarsson S. Molecular Diversity of Midbrain Development in Mouse, Human, and Stem Cells. Cell 2017; 167:566-580.e19. [PMID: 27716510 PMCID: PMC5055122 DOI: 10.1016/j.cell.2016.09.027] [Citation(s) in RCA: 572] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 07/14/2016] [Accepted: 09/16/2016] [Indexed: 12/11/2022]
Abstract
Understanding human embryonic ventral midbrain is of major interest for Parkinson's disease. However, the cell types, their gene expression dynamics, and their relationship to commonly used rodent models remain to be defined. We performed single-cell RNA sequencing to examine ventral midbrain development in human and mouse. We found 25 molecularly defined human cell types, including five subtypes of radial glia-like cells and four progenitors. In the mouse, two mature fetal dopaminergic neuron subtypes diversified into five adult classes during postnatal development. Cell types and gene expression were generally conserved across species, but with clear differences in cell proliferation, developmental timing, and dopaminergic neuron development. Additionally, we developed a method to quantitatively assess the fidelity of dopaminergic neurons derived from human pluripotent stem cells, at a single-cell level. Thus, our study provides insight into the molecular programs controlling human midbrain development and provides a foundation for the development of cell replacement therapies.
Collapse
Affiliation(s)
- Gioele La Manno
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Science for Life Laboratory, 17121 Solna, Sweden
| | - Daniel Gyllborg
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Simone Codeluppi
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Science for Life Laboratory, 17121 Solna, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Kaneyasu Nishimura
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Carmen Salto
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Amit Zeisel
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Science for Life Laboratory, 17121 Solna, Sweden
| | - Lars E Borm
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Science for Life Laboratory, 17121 Solna, Sweden
| | - Simon R W Stott
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Enrique M Toledo
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - J Carlos Villaescusa
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Psychiatric Stem Cell Group, Neurogenetics Unit, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Peter Lönnerberg
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Science for Life Laboratory, 17121 Solna, Sweden
| | - Jesper Ryge
- Laboratory of Neural Microcircuitry, Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Ernest Arenas
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.
| | - Sten Linnarsson
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Science for Life Laboratory, 17121 Solna, Sweden.
| |
Collapse
|
35
|
Yang L, Jiang Z, Zhou L, Zhao K, Ma X, Cheng G. Hydrophilic cell-derived extracellular matrix as a niche to promote adhesion and differentiation of neural progenitor cells. RSC Adv 2017. [DOI: 10.1039/c7ra08273h] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cell-derived extracellular matrix exhibits excellent adhesion performance for neural progenitor cell anchoring and residency, resulting in promoted proliferation of the stem cells to basal forebrain cholinergic neurons.
Collapse
Affiliation(s)
- Lingyan Yang
- CAS Key Laboratory of Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- China
- School of Nano Technology and Nano Bionics
| | - Ziyun Jiang
- CAS Key Laboratory of Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- China
| | - Linhong Zhou
- Department of Pharmacy
- School of Medicine
- Xi'an Jiaotong University
- China
| | - Keli Zhao
- CAS Key Laboratory of Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- China
| | - Xun Ma
- CAS Key Laboratory of Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- China
| | - Guosheng Cheng
- CAS Key Laboratory of Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- China
- School of Nano Technology and Nano Bionics
| |
Collapse
|
36
|
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: 3.1] [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]
|
37
|
Abstract
Stem cells hold great promise in treating many diseases either through promoting endogenous cell repair or through direct cell transplants. In order to maximize their potential, understanding the fundamental signals and mechanisms that regulate their behavior is essential. The extracellular matrix (ECM) is one such component involved in mediating stem cell fate. Recent studies have made significant progress in understanding stem cell-ECM interactions. Technological developments have provided greater clarity in how cells may sense and respond to the ECM, in particular the physical properties of the matrix. This review summarizes recent developments, providing illustrative examples of the different modes with which the ECM controls both embryonic and adult stem cell behavior.
Collapse
|
38
|
Safina D, Schlitt F, Romeo R, Pflanzner T, Pietrzik CU, Narayanaswami V, Edenhofer F, Faissner A. Low-density lipoprotein receptor-related protein 1 is a novel modulator of radial glia stem cell proliferation, survival, and differentiation. Glia 2016; 64:1363-80. [PMID: 27258849 DOI: 10.1002/glia.23009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 04/19/2016] [Accepted: 05/04/2016] [Indexed: 12/22/2022]
Abstract
The LDL family of receptors and its member low-density lipoprotein receptor-related protein 1 (LRP1) have classically been associated with a modulation of lipoprotein metabolism. Current studies, however, indicate diverse functions for this receptor in various aspects of cellular activities, including cell proliferation, migration, differentiation, and survival. LRP1 is essential for normal neuronal function in the adult CNS, whereas the role of LRP1 in development remained unclear. Previously, we have observed an upregulation of LewisX (LeX) glycosylated LRP1 in the stem cells of the developing cortex and demonstrated its importance for oligodendrocyte differentiation. In the current study, we show that LeX-glycosylated LRP1 is also expressed in the stem cell compartment of the developing spinal cord and has broader functions in the developing CNS. We have investigated the basic properties of LRP1 conditional knockout on the neural stem/progenitor cells (NSPCs) from the cortex and the spinal cord, created by means of Cre-loxp-mediated recombination in vitro. The functional status of LRP1-deficient cells has been studied using proliferation, differentiation, and apoptosis assays. LRP1 deficient NSPCs from both CNS regions demonstrated altered differentiation profiles. Their differentiation capacity toward oligodendrocyte progenitor cells (OPCs), mature oligodendrocytes and neurons was reduced. In contrast, astrocyte differentiation was promoted. Moreover, LRP1 deletion had a negative effect on NSPCs proliferation and survival. Our observations suggest that LRP1 facilitates NSPCs differentiation via interaction with apolipoprotein E (ApoE). Upon ApoE4 stimulation wild type NSPCs generated more oligodendrocytes, but LRP1 knockout cells showed no response. The effect of ApoE seems to be independent of cholesterol uptake, but is rather mediated by downstream MAPK and Akt activation. GLIA 2016 GLIA 2016;64:1363-1380.
Collapse
Affiliation(s)
- Dina Safina
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University, Bochum, D-44780, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, D-44780, Germany
| | - Frederik Schlitt
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University, Bochum, D-44780, Germany
| | - Ramona Romeo
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University, Bochum, D-44780, Germany
| | - Thorsten Pflanzner
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, D-55099, Germany
| | - Claus U Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, D-55099, Germany
| | - Vasanthy Narayanaswami
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, California, 90840
| | - Frank Edenhofer
- Institute of Anatomy and Cell Biology, University Wuerzburg, Koellikerstraße 6, Wuerzburg, D-97070, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University, Bochum, D-44780, Germany
| |
Collapse
|
39
|
Reinhard J, Brösicke N, Theocharidis U, Faissner A. The extracellular matrix niche microenvironment of neural and cancer stem cells in the brain. Int J Biochem Cell Biol 2016; 81:174-183. [PMID: 27157088 DOI: 10.1016/j.biocel.2016.05.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/25/2016] [Accepted: 05/04/2016] [Indexed: 12/27/2022]
Abstract
Numerous studies demonstrated that neural stem cells and cancer stem cells (NSCs/CSCs) share several overlapping characteristics such as self-renewal, multipotency and a comparable molecular repertoire. In addition to the intrinsic cellular properties, NSCs/CSCs favor a similar environment to acquire and maintain their characteristics. In the present review, we highlight the shared properties of NSCs and CSCs in regard to their extracellular microenvironment called the NSC/CSC niche. Moreover, we point out that extracellular matrix (ECM) molecules and their complementary receptors influence the behavior of NSCs/CSCs as well as brain tumor progression. Here, we focus on the expression profile and functional importance of the ECM glycoprotein tenascin-C, the chondroitin sulfate proteoglycan DSD-1-PG/phosphacan but also on other important glycoprotein/proteoglycan constituents. Within this review, we specifically concentrate on glioblastoma multiforme (GBM). GBM is the most common malignant brain tumor in adults and is associated with poor prognosis despite intense and aggressive surgical and therapeutic treatment. Recent studies indicate that GBM onset is driven by a subpopulation of CSCs that display self-renewal and recapitulate tumor heterogeneity. Based on the CSC hypothesis the cancer arises just from a small subpopulation of self-sustaining cancer cells with the exclusive ability to self-renew and maintain the tumor. Besides the fundamental stem cell properties of self-renewal and multipotency, GBM stem cells share further molecular characteristics with NSCs, which we would like to review in this article.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Nicole Brösicke
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Ursula Theocharidis
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany.
| |
Collapse
|
40
|
Regalado-Santiago C, Juárez-Aguilar E, Olivares-Hernández JD, Tamariz E. Mimicking Neural Stem Cell Niche by Biocompatible Substrates. Stem Cells Int 2016; 2016:1513285. [PMID: 26880934 PMCID: PMC4736764 DOI: 10.1155/2016/1513285] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/19/2015] [Accepted: 11/23/2015] [Indexed: 01/17/2023] Open
Abstract
Neural stem cells (NSCs) participate in the maintenance, repair, and regeneration of the central nervous system. During development, the primary NSCs are distributed along the ventricular zone of the neural tube, while, in adults, NSCs are mainly restricted to the subependymal layer of the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus in the hippocampus. The circumscribed areas where the NSCs are located contain the secreted proteins and extracellular matrix components that conform their niche. The interplay among the niche elements and NSCs determines the balance between stemness and differentiation, quiescence, and proliferation. The understanding of niche characteristics and how they regulate NSCs activity is critical to building in vitro models that include the relevant components of the in vivo niche and to developing neuroregenerative approaches that consider the extracellular environment of NSCs. This review aims to examine both the current knowledge on neurogenic niche and how it is being used to develop biocompatible substrates for the in vitro and in vivo mimicking of extracellular NSCs conditions.
Collapse
Affiliation(s)
- Citlalli Regalado-Santiago
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Enrique Juárez-Aguilar
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Juan David Olivares-Hernández
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Elisa Tamariz
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| |
Collapse
|
41
|
Wiese S, Faissner A. The role of extracellular matrix in spinal cord development. Exp Neurol 2015; 274:90-9. [DOI: 10.1016/j.expneurol.2015.05.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/13/2015] [Accepted: 05/25/2015] [Indexed: 01/06/2023]
|
42
|
Karus M, Ulc A, Ehrlich M, Czopka T, Hennen E, Fischer J, Mizhorova M, Qamar N, Brüstle O, Faissner A. Regulation of oligodendrocyte precursor maintenance by chondroitin sulphate glycosaminoglycans. Glia 2015; 64:270-86. [PMID: 26454153 DOI: 10.1002/glia.22928] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 09/16/2015] [Indexed: 01/06/2023]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) have been proven to inhibit morphological maturation of oligodendrocytes as well as their myelination capabilities. Yet, it remained unclear, whether CSPGs and/or their respective chondroitin sulfate glycosaminoglycan (CS-GAG) side chains also regulate the oligodendrocyte lineage progression. Here, we initially show that CS-GAGs detected by the monoclonal antibody 473HD are expressed by primary rat NG2-positive oligodendrocyte precursor cells (OPCs) and O4-positive immature oligodendrocytes. CS-GAGs become down-regulated with ongoing oligodendrocyte differentiation. Enzymatic removal of the CS-GAG chains by the bacterial enzyme Chondroitinase ABC (ChABC) promoted spontaneous differentiation of proliferating rat OPCs toward O4-positive immature oligodendrocytes. Upon forced differentiation, the enzymatic removal of the CS-GAGs accelerated oligodendrocyte differentiation toward both MBP-positive and membrane forming oligodendrocytes. These processes were attenuated on enriched CSPG fractions, mainly consisting of Phosphacan/RPTPβ/ζ and to less extent of Brevican and NG2. To qualify CS-GAGs as universal regulators of oligodendrocyte biology, we finally tested the effect of CS-GAG removal on OPCs from different sources such as mouse cortical oligospheres, mouse spinal cord neurospheres, and most importantly human-induced pluripotent stem cell-derived radial glia-like neural precursor cells. For all culture systems used, we observed a similar inhibitory effect of CS-GAGs on oligodendrocyte differentiation. In conclusion, this study clearly suggests an important fundamental principle for complex CS-GAGs to regulate the oligodendrocyte lineage progression. Moreover, the use of ChABC in order to promote oligodendrocyte differentiation toward myelin gene expressing cells might be an applicable therapeutic option to enhance white matter repair.
Collapse
Affiliation(s)
- Michael Karus
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany.,Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, Bonn, Germany
| | - Annika Ulc
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Marc Ehrlich
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Tim Czopka
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Eva Hennen
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Julia Fischer
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, Bonn, Germany
| | - Marija Mizhorova
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, Bonn, Germany
| | - Naila Qamar
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, Bonn, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
43
|
Faissner A, Reinhard J. The extracellular matrix compartment of neural stem and glial progenitor cells. Glia 2015; 63:1330-49. [DOI: 10.1002/glia.22839] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/25/2015] [Accepted: 03/30/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology; Ruhr-University Bochum; Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology; Ruhr-University Bochum; Germany
| |
Collapse
|
44
|
Tsai HL, Chiu WT, Fang CL, Hwang SM, Renshaw PF, Lai WFT. Different forms of tenascin-C with tenascin-R regulate neural differentiation in bone marrow-derived human mesenchymal stem cells. Tissue Eng Part A 2015; 20:1908-21. [PMID: 24829055 DOI: 10.1089/ten.tea.2013.0188] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are currently thought to transdifferentiate into neural lineages under specific microenvironments. Studies have reported that the tenascin family members, tenascin-C (TnC) and tenascin-R (TnR), regulate differentiation and migration, in addition to neurite outgrowth and survival in numerous types of neurons and mesenchymal progenitor cells. However, the mechanisms by which TnC and TnR affect neuronal differentiation are not well understood. In this study, we hypothesized that different forms of tenascin might regulate the neural transdifferentiation of human bone marrow-derived mesenchymal stem cells. Human MSCs were cultured in media incorporated with soluble tenascins, or on precoated tenascins. In a qualitative polymerase chain reaction analysis, adding a soluble TnC and TnR mixture to the medium significantly enhanced the expression of neuronal and glial markers, whereas no synaptic markers were expressed. Conversely, in groups of cells treated with coated TnC, hMSCs showed neurite outgrowth and synaptic marker expression. After being treated with coated TnR, hMSCs exhibited neuronal differentiation; however, it inhibited neurite outgrowth and synaptic marker expression. A combination of TnC and TnR significantly promoted hMSC differentiation in neurons or oligodendrocytes, induced neurite formation, and inhibited differentiation into astrocytes. Furthermore, the effect of the tenascin mixture showed dose-dependent effects, and a mixture ratio of 1:1 to 1:2 (TnC:TnR) provided the most obvious differentiation of neurons and oligodendrocytes. In a functional blocking study, integrin α7 and α9β1-blocking antibodies inhibited, respectively, 80% and 20% of mRNA expression by hMSCs in the coated tenascin mixture. In summary, the coated combination of TnC and TnR appeared to regulate neural differentiation signaling through integrin α7 and α9β1 in bone marrow-derived hMSCs. Our findings demonstrate novel mechanisms by which tenascin regulates neural differentiation, and enable the use of cell therapy to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Hung-Li Tsai
- 1 Graduate Institute of Medical Sciences, Taipei Medical University , Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
45
|
The guanine nucleotide exchange factor Vav3 regulates differentiation of progenitor cells in the developing mouse retina. Cell Tissue Res 2014; 359:423-440. [PMID: 25501893 DOI: 10.1007/s00441-014-2050-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 11/03/2014] [Indexed: 10/24/2022]
Abstract
The seven main cell types in the mammalian retina arise from multipotent retinal progenitor cells, a process that is tightly regulated by intrinsic and extrinsic signals. However, the molecular mechanisms that control proliferation, differentiation and cell-fate decisions of retinal progenitor cells are not fully understood yet. Here, we report that the guanine nucleotide exchange factor Vav3, a regulator of Rho-GTPases, is involved in retinal development. We demonstrate that Vav3 is expressed in the mouse retina during the embryonic period. In order to study the role of Vav3 in the developing retina, we generate Vav3-deficient mice. The loss of Vav3 results in an accelerated differentiation of retinal ganglion cells and cone photoreceptors during early and late embryonic development. We provide evidence that more retinal progenitor cells express the late progenitor marker Sox9 in Vav3-deficient mice than in wild-types. This premature differentiation is compensated during the postnatal period and late-born cell types such as bipolar cells and Müller glia display normal numbers. Taken together, our data imply that Vav3 is a regulator of retinal progenitor cell differentiation, thus highlighting a novel role for guanine nucleotide exchange factors in retinogenesis.
Collapse
|
46
|
Hendaoui I, Tucker RP, Zingg D, Bichet S, Schittny J, Chiquet-Ehrismann R. Tenascin-C is required for normal Wnt/β-catenin signaling in the whisker follicle stem cell niche. Matrix Biol 2014; 40:46-53. [PMID: 25196097 DOI: 10.1016/j.matbio.2014.08.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 01/01/2023]
Abstract
Whisker follicles have multiple stem cell niches, including epidermal stem cells in the bulge as well as neural crest-derived stem cells and mast cell progenitors in the trabecular region. The neural crest-derived stem cells are a pool of melanocyte precursors. Previously, we found that the extracellular matrix glycoproteins tenascin-C and tenascin-W are expressed near CD34-positive cells in the trabecular stem cell niche of mouse whisker follicles. Here, we analyzed whiskers from tenascin-C knockout mice and found intrafollicular adipocytes and supernumerary mast cells. As Wnt/β-catenin signaling promotes melanogenesis and suppresses the differentiation of adipocytes and mast cells, we analyzed β-catenin subcellular localization in the trabecular niche. We found cytoplasmic and nuclear β-catenin in wild-type mice reflecting active Wnt/β-catenin signaling, whereas β-catenin in tenascin-C knockout mice was mostly cell membrane-associated and thus transcriptionally inactive. Furthermore, cells expressing the Wnt/β-catenin target gene cyclin D1 were enriched in the CD34-positive niches of wild-type compared to tenascin-C knockout mice. We then tested the effects of tenascins on this signaling pathway. We found that tenascin-C and tenascin-W can be co-precipitated with Wnt3a. In vitro, substrate bound tenascins promoted β-catenin-mediated transcription in the presence of Wnt3a, presumably due to the sequestration and concentration of Wnt3a near the cell surface. We conclude that the presence of tenascin-C in whiskers assures active Wnt/β-catenin signaling in the niche thereby maintaining the stem cell pool and suppressing aberrant differentiation, while in the knockout mice with reduced Wnt/β-catenin signaling, stem cells from the trabecular niche can differentiate into ectopic adipocytes and mast cells.
Collapse
Affiliation(s)
- Ismaïl Hendaoui
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, Basel, Switzerland
| | - Richard P Tucker
- Department of Cell Biology and Human Anatomy, University of California at Davis, Davis, CA, USA
| | - Dominik Zingg
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, Basel, Switzerland
| | - Sandrine Bichet
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, Basel, Switzerland
| | | | - Ruth Chiquet-Ehrismann
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, Basel, Switzerland; Faculty of Science, University of Basel, Basel, Switzerland.
| |
Collapse
|
47
|
Ikeshima-Kataoka H, Abe Y, Yasui M. Aquaporin 4-dependent expression of glial fibrillary acidic protein and tenascin-C in activated astrocytes in stab wound mouse brain and in primary culture. J Neurosci Res 2014; 93:121-9. [PMID: 25174305 DOI: 10.1002/jnr.23467] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/23/2014] [Accepted: 07/11/2014] [Indexed: 01/14/2023]
Abstract
We previously reported that aquaporin 4 (AQP4) has a neuroimmunological function via astrocytes and microglial cells involving osteopontin. AQP4 is a water channel localized in the endofoot of astrocytes in the brain, and its expression is upregulated after a stab wound to the mouse brain or the injection of methylmercury in common marmosets. In this study, the correlation between the expression of AQP4 and the expression of glial fibrillary acidic protein (GFAP) or tenascin-C (TN-C) in reactive astrocytes was examined in primary cultures and brain tissues of AQP4-deficient mice (AQP4/KO). In the absence of a stab wound to the brain or of any stimulation of the cells, the expressions of both GFAP and TN-C were lower in astrocytes from AQP4/KO mice than in those from wild-type (WT) mice. High levels of GFAP and TN-C expression were observed in activated astrocytes after a stab wound to the brain in WT mice; however, the expressions of GFAP and TN-C were insignificant in AQP4/KO mice. Furthermore, lipopolysaccharide (LPS) stimulation activated primary culture of astrocytes and upregulated GFAP and TN-C expression in cells from WT mice, whereas the expressions of GFAP and TN-C were slightly upregulated in cells from AQP4/KO mice. Moreover, the stimulation of primary culture of astrocytes with LPS also upregulated inflammatory cytokines in cells from WT mice, whereas modest increases were observed in cells from AQP4/KO mice. These results suggest that AQP4 expression accelerates GFAP and TN-C expression in activated astrocytes induced by a stab wound in the mouse brain and LPS-stimulated primary culture of astrocytes.
Collapse
Affiliation(s)
- Hiroko Ikeshima-Kataoka
- Department of Pharmacology and Neuroscience, Keio University School of Medicine, Tokyo, Japan; Faculty of Science and Engineering, Waseda University, Tokyo, Japan
| | | | | |
Collapse
|
48
|
Roll L, Faissner A. Influence of the extracellular matrix on endogenous and transplanted stem cells after brain damage. Front Cell Neurosci 2014; 8:219. [PMID: 25191223 PMCID: PMC4137450 DOI: 10.3389/fncel.2014.00219] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/18/2014] [Indexed: 01/07/2023] Open
Abstract
The limited regeneration capacity of the adult central nervous system (CNS) requires strategies to improve recovery of patients. In this context, the interaction of endogenous as well as transplanted stem cells with their environment is crucial. An understanding of the molecular mechanisms could help to improve regeneration by targeted manipulation. In the course of reactive gliosis, astrocytes upregulate Glial fibrillary acidic protein (GFAP) and start, in many cases, to proliferate. Beside GFAP, subpopulations of these astroglial cells coexpress neural progenitor markers like Nestin. Although cells express these markers, the proportion of cells that eventually give rise to neurons is limited in many cases in vivo compared to the situation in vitro. In the first section, we present the characteristics of endogenous progenitor-like cells and discuss the differences in their neurogenic potential in vitro and in vivo. As the environment plays an important role for survival, proliferation, migration, and other processes, the second section of the review describes changes in the extracellular matrix (ECM), a complex network that contains numerous signaling molecules. It appears that signals in the damaged CNS lead to an activation and de-differentiation of astrocytes, but do not effectively promote neuronal differentiation of these cells. Factors that influence stem cells during development are upregulated in the damaged brain as part of an environment resembling a stem cell niche. We give a general description of the ECM composition, with focus on stem cell-associated factors like the glycoprotein Tenascin-C (TN-C). Stem cell transplantation is considered as potential treatment strategy. Interaction of transplanted stem cells with the host environment is critical for the outcome of stem cell-based therapies. Possible mechanisms involving the ECM by which transplanted stem cells might improve recovery are discussed in the last section.
Collapse
Affiliation(s)
- Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany ; International Graduate School of Neuroscience, Ruhr-University Bochum Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany ; International Graduate School of Neuroscience, Ruhr-University Bochum Bochum, Germany
| |
Collapse
|
49
|
Chiquet-Ehrismann R, Orend G, Chiquet M, Tucker RP, Midwood KS. Tenascins in stem cell niches. Matrix Biol 2014; 37:112-23. [DOI: 10.1016/j.matbio.2014.01.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/15/2014] [Accepted: 01/16/2014] [Indexed: 12/16/2022]
|
50
|
Karus M, Blaess S, Brüstle O. Self-organization of neural tissue architectures from pluripotent stem cells. J Comp Neurol 2014; 522:2831-44. [PMID: 24737617 DOI: 10.1002/cne.23608] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 12/23/2022]
Abstract
Despite being a subject of intensive research, the mechanisms underlying the formation of neural tissue architectures during development of the central nervous system remain largely enigmatic. So far, studies into neural pattern formation have been restricted mainly to animal experiments. With the advent of pluripotent stem cells it has become possible to explore early steps of nervous system development in vitro. These studies have unraveled a remarkable propensity of primitive neural cells to self-organize into primitive patterns such as neural tube-like rosettes in vitro. Data from more advanced 3D culture systems indicate that this intrinsic propensity for self-organization can even extend to the formation of complex architectures such as a multilayered cortical neuroepithelium or an entire optic cup. These novel experimental paradigms not only demonstrate the enormous self-organization capacity of neural stem cells, they also provide exciting prospects for studying the earliest steps of human neural tissue development and the pathogenesis of brain malformations in reductionist in vitro paradigms.
Collapse
Affiliation(s)
- Michael Karus
- Institute of Reconstructive Neurobiology, University of Bonn LIFE&BRAIN Center, and LIFE&BRAIN GmbH, 53127, Bonn, Germany
| | | | | |
Collapse
|