1
|
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
|
2
|
Chen W, Wu Y, Wang J, Yu W, Shen X, Zhao K, Liang B, Hu X, Wang S, Jiang H, Liu X, Zhang M, Xing X, Wang C, Xing D. Clinical advances in TNC delivery vectors and their conjugate agents. Pharmacol Ther 2024; 253:108577. [PMID: 38081519 DOI: 10.1016/j.pharmthera.2023.108577] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
Tenascin C (TNC), a glycoprotein that is abundant in the tumor extracellular matrix (ECM), is strongly overexpressed in tumor tissues but virtually undetectable in most normal tissues. Many TNC antibodies, peptides, aptamers, and nanobodies have been investigated as delivery vectors, including 20A1, α-A2, α-A3, α-IIIB, α-D, BC-2, BC-4 BC-8, 81C6, ch81C6, F16, FHK, Ft, Ft-NP, G11, G11-iRGD, GBI-10, 19H12, J1/TN1, J1/TN2, J1/TN3, J1/TN4, J1/TN5, NJT3, NJT4, NJT6, P12, PL1, PL3, R6N, SMART, ST2146, ST2485, TN11, TN12, TNFnA1A2-Fc, TNfnA1D-Fc, TNfnBD-Fc, TNFnCD-Fc, TNfnD6-Fc, TNfn78-Fc, TTA1, TTA1.1, and TTA1.2. In particular, BC-2, BC-4, 81C6, ch81C6, F16, FHK, G11, PL1, PL3, R6N, ST2146, TN11, and TN12 have been tested in human tissues. G11-iRGD and simultaneous multiple aptamers and arginine-glycine-aspartic acid (RGD) targeting (SMART) may be assessed in clinical trials because G11, iRGD and AS1411 (SMART components) are already in clinical trials. Many TNC-conjugate agents, including antibody-drug conjugates (ADCs), antibody fragment-drug conjugates (FDCs), immune-stimulating antibody conjugates (ISACs), and radionuclide-drug conjugates (RDCs), have been investigated in preclinical and clinical trials. RDCs investigated in clinical trials include 111In-DTPA-BC-2, 131I-BC-2, 131I-BC-4, 90Y-BC4, 131I81C6, 131I-ch81C6, 211At-ch81C6, F16124I, 131I-tenatumomab, ST2146biot, FDC 131I-F16S1PF(ab')2, and ISAC F16IL2. ADCs (including FHK-SSL-Nav, FHK-NB-DOX, Ft-NP-PTX, and F16*-MMAE) and ISACs (IL12-R6N and 125I-G11-IL2) may enter clinical trials because they contain components of marketed treatments or agents that were investigated in previous clinical studies. This comprehensive review presents historical perspectives on clinical advances in TNC-conjugate agents to provide timely information to facilitate tumor-targeting drug development using TNC.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Jie Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Wanpeng Yu
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Xin Shen
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, Shandong 266042, China
| | - Kai Zhao
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China; Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Bing Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Xiaokun Hu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China; Interventional Medicine Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Shuai Wang
- Department of Radiotherapy, Affiliated Hospital of Weifang Medical University, Key Laboratory of Precision Radiation Therapy for Tumors in Weifang City, School of Medical Imaging, Weifang Medical University, Weifang, Shandong 261031, China
| | - Hongfei Jiang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Xinlin Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Miao Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Xiaohui Xing
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China.
| | - Chao Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
3
|
Gschwandtner M, Gammage AN, Deligne C, Mies LFM, Domaingo A, Murdamoothoo D, Loustau T, Schwenzer A, Derler R, Carapito R, Koch M, Mörgelin M, Orend G, Kungl AJ, Midwood KS. Investigating Chemokine-Matrix Networks in Breast Cancer: Tenascin-C Sets the Tone for CCL2. Int J Mol Sci 2023; 24:8365. [PMID: 37176074 PMCID: PMC10179296 DOI: 10.3390/ijms24098365] [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: 03/31/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Bidirectional dialogue between cellular and non-cellular components of the tumor microenvironment (TME) drives cancer survival. In the extracellular space, combinations of matrix molecules and soluble mediators provide external cues that dictate the behavior of TME resident cells. Often studied in isolation, integrated cues from complex tissue microenvironments likely function more cohesively. Here, we study the interplay between the matrix molecule tenascin-C (TNC) and chemokine CCL2, both elevated in and associated with the progression of breast cancer and playing key roles in myeloid immune responses. We uncover a correlation between TNC/CCL2 tissue levels in HER2+ breast cancer and examine the physical and functional interactions of these molecules in a murine disease model with tunable TNC levels and in in vitro cellular and cell-free models. TNC supported sustained CCL2 synthesis, with chemokine binding to TNC via two distinct domains. TNC dominated the behavior of tumor-resident myeloid cells; CCL2 did not impact macrophage survival/activation whilst TNC facilitated an immune suppressive macrophage phenotype that was not dependent on or altered by CCL2 co-expression. Together, these data map new binding partners within the TME and demonstrate that whilst the matrix exerts transcriptional control over the chemokine, each plays a distinct role in subverting anti-tumoral immunity.
Collapse
Affiliation(s)
| | - Anís N. Gammage
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Claire Deligne
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Linda F. M. Mies
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Alissa Domaingo
- Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Devardarssen Murdamoothoo
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, 67091 Strasbourg, France
- University of Strasbourg, 67091 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67091 Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, 67091 Strasbourg, France
| | - Thomas Loustau
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, 67091 Strasbourg, France
- University of Strasbourg, 67091 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67091 Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, 67091 Strasbourg, France
| | - Anja Schwenzer
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Rupert Derler
- Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Raphael Carapito
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67091 Strasbourg, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, GENOMAX Platform, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, 67091 Strasbourg, France
| | - Manuel Koch
- Institute for Dental Research and Oral, Musculoskeletal Research, Center for Biochemistry, University of Cologne, 50931 Cologne, Germany
| | | | - Gertraud Orend
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, 67091 Strasbourg, France
- University of Strasbourg, 67091 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67091 Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, 67091 Strasbourg, France
| | - Andreas J. Kungl
- Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Kim S. Midwood
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| |
Collapse
|
4
|
Bijelić D, Adžić M, Perić M, Reiss G, Milošević M, Andjus PR, Jakovčevski I. Tenascin-C fibronectin D domain is involved in the fine-tuning of glial response to CNS injury in vitro. Front Cell Dev Biol 2022; 10:952208. [PMID: 36092707 PMCID: PMC9462431 DOI: 10.3389/fcell.2022.952208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/18/2022] [Indexed: 11/19/2022] Open
Abstract
Understanding processes that occur after injuries to the central nervous system is essential in order to gain insight into how the restoration of function can be improved. Extracellular glycoprotein tenascin-C (TnC) has numerous functions in wound healing process depending on the expression time, location, isoform and binding partners which makes it interesting to study in this context. We used an in vitro injury model, the mixed culture of cortical astrocytes and microglia, and observed that without TnC microglial cells tend to populate gap area in greater numbers and proliferate more, whereas astrocytes build up in the border region to promote faster gap closure. Alternatively spliced domain of TnC, fibronectin type III-like repeat D (FnD) strongly affected physiological properties and morphology of both astrocytes and microglia in this injury model. The rate of microglial proliferation in the injury region decreased significantly with the addition of FnD. Additionally, density of microglia also decreased, in part due to reduced proliferation, and possibly due to reduced migration and increased contact inhibition between enlarged FnD-treated cells. Overall morphology of FnD-treated microglia resembled the activated pro-inflammatory cells, and elevated expression of iNOS was in accordance with this phenotype. The effect of FnD on astrocytes was different, as it did not affect their proliferation, but stimulated migration of reactivated astrocytes into the scratched area 48 h after the lesion. Elevated expression and secretion of TNF-α and IL-1β upon FnD treatment indicated the onset of inflammation. Furthermore, on Western blots we observed increased intensity of precursor bands of β1 integrin and appearance of monomeric bands of P2Y12R after FnD treatment which substantiates and clarifies its role in cellular shape and motility changes. Our results show versatile functions of TnC and in particular FnD after injury, mostly contributing to ongoing inflammation in the injury region. Based on our findings, FnD might be instrumental in limiting immune cell infiltration, and promoting astrocyte migration within the injury region, thus influencing spaciotemporal organization of the wound and surrounding area.
Collapse
Affiliation(s)
- Dunja Bijelić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- *Correspondence: Dunja Bijelić, ; Igor Jakovčevski,
| | - Marija Adžić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Mina Perić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Gebhard Reiss
- Institute for Anatomy and Clinical Morphology, University Witten / Herdecke, Witten, Germany
| | - Milena Milošević
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Pavle R. Andjus
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Igor Jakovčevski
- Institute for Anatomy and Clinical Morphology, University Witten / Herdecke, Witten, Germany
| |
Collapse
|
5
|
Structural and Functional Modulation of Perineuronal Nets: In Search of Important Players with Highlight on Tenascins. Cells 2021; 10:cells10061345. [PMID: 34072323 PMCID: PMC8230358 DOI: 10.3390/cells10061345] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) of the brain plays a crucial role in providing optimal conditions for neuronal function. Interactions between neurons and a specialized form of ECM, perineuronal nets (PNN), are considered a key mechanism for the regulation of brain plasticity. Such an assembly of interconnected structural and regulatory molecules has a prominent role in the control of synaptic plasticity. In this review, we discuss novel ways of studying the interplay between PNN and its regulatory components, particularly tenascins, in the processes of synaptic plasticity, mechanotransduction, and neurogenesis. Since enhanced neuronal activity promotes PNN degradation, it is possible to study PNN remodeling as a dynamical change in the expression and organization of its constituents that is reflected in its ultrastructure. The discovery of these subtle modifications is enabled by the development of super-resolution microscopy and advanced methods of image analysis.
Collapse
|
6
|
Giblin SP, Schwenzer A, Midwood KS. Alternative splicing controls cell lineage-specific responses to endogenous innate immune triggers within the extracellular matrix. Matrix Biol 2020; 93:95-114. [PMID: 32599145 DOI: 10.1016/j.matbio.2020.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/15/2020] [Accepted: 06/21/2020] [Indexed: 01/08/2023]
Abstract
The identification of barely more than 20,000 human genes was amongst the most surprising outcomes of the human genome project. Alternative splicing provides an essential means of expanding the proteome, enabling a single gene to encode multiple, distinct isoforms by selective inclusion or exclusion of exons from mature mRNA. However, mis-regulation of this process is associated with most human diseases. Here, we examine the impact of post-transcriptional processing on extracellular matrix function, focusing on the complex alternative splicing patterns of tenascin-C, a molecule that can exist in as many as 500 different isoforms. We demonstrate that the pro-inflammatory activity of this endogenous innate immune trigger is controlled by inclusion or exclusion of a novel immunomodulatory site located within domains AD2AD1, identifying this as a mechanism that prevents unnecessary inflammation in healthy tissues but enables rapid immune cell mobilization and activation upon tissue damage, and defining how this goes awry in autoimmune disease.
Collapse
Affiliation(s)
- Sean P Giblin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Anja Schwenzer
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Kim S Midwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
7
|
Ponroy Bally B, Farmer WT, Jones EV, Jessa S, Kacerovsky JB, Mayran A, Peng H, Lefebvre JL, Drouin J, Hayer A, Ernst C, Murai KK. Human iPSC-derived Down syndrome astrocytes display genome-wide perturbations in gene expression, an altered adhesion profile, and increased cellular dynamics. Hum Mol Genet 2020; 29:785-802. [PMID: 31943018 PMCID: PMC7104679 DOI: 10.1093/hmg/ddaa003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/31/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Down syndrome (DS), caused by the triplication of human chromosome 21, leads to significant alterations in brain development and is a major genetic cause of intellectual disability. While much is known about changes to neurons in DS, the effects of trisomy 21 on non-neuronal cells such as astrocytes are poorly understood. Astrocytes are critical for brain development and function, and their alteration may contribute to DS pathophysiology. To better understand the impact of trisomy 21 on astrocytes, we performed RNA-sequencing on astrocytes from newly produced DS human induced pluripotent stem cells (hiPSCs). While chromosome 21 genes were upregulated in DS astrocytes, we found consistent up- and down-regulation of genes across the genome with a strong dysregulation of neurodevelopmental, cell adhesion and extracellular matrix molecules. ATAC (assay for transposase-accessible chromatin)-seq also revealed a global alteration in chromatin state in DS astrocytes, showing modified chromatin accessibility at promoters of cell adhesion and extracellular matrix genes. Along with these transcriptomic and epigenomic changes, DS astrocytes displayed perturbations in cell size and cell spreading as well as modifications to cell-cell and cell-substrate recognition/adhesion, and increases in cellular motility and dynamics. Thus, triplication of chromosome 21 is associated with genome-wide transcriptional, epigenomic and functional alterations in astrocytes that may contribute to altered brain development and function in DS.
Collapse
Affiliation(s)
- Blandine Ponroy Bally
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - W Todd Farmer
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - Emma V Jones
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - Selin Jessa
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
- Quantitative Life Sciences, McGill University, Montreal, QC H3A 2A7, Canada
| | - J Benjamin Kacerovsky
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - Alexandre Mayran
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Huashan Peng
- Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
- Department of Human Genetics, McGill University, Montreal, QC H4H 1R3, Canada
- Douglas Hospital Research Institute, Verdun, QC H4H 1R3, Canada
| | - Julie L Lefebvre
- Department of Molecular Genetics, Program for Neuroscience and Mental Health, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Jacques Drouin
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Arnold Hayer
- Department of Biology, McGill University, Bellini Life Sciences Complex, Montreal, QC H3G 0B1, Canada
| | - Carl Ernst
- Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
- Department of Human Genetics, McGill University, Montreal, QC H4H 1R3, Canada
- Douglas Hospital Research Institute, Verdun, QC H4H 1R3, Canada
| | - Keith K Murai
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
- Quantitative Life Sciences, McGill University, Montreal, QC H3A 2A7, Canada
| |
Collapse
|
8
|
Reginensi D, Ortiz D, Pravia A, Burillo A, Morales F, Morgan C, Jimenez L, Dave KR, Perez-Pinzon MA, Gittens RA. Role of Region-Specific Brain Decellularized Extracellular Matrix on In Vitro Neuronal Maturation. Tissue Eng Part A 2020; 26:964-978. [PMID: 32103711 DOI: 10.1089/ten.tea.2019.0277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recent advancements in tissue engineering suggest that biomaterials, such as decellularized extracellular matrix (ECM), could serve to potentiate the localization and efficacy of regenerative therapies in the central nervous system. Still, what factors and which mechanisms are required from these ECM-based biomaterials to exert their effect are not entirely understood. In this study, we use the brain as a novel model to test the effects of particular biochemical and structural properties by evaluating, for the first time, three different sections of the brain (i.e., cortex, cerebellum, and remaining areas) side-by-side and their corresponding decellularized counterparts using mechanical (4-day) and chemical (1-day) decellularization protocols. The three different brain subregions had considerably different initial conditions in terms of cell number and growth factor content, and some of these differences were maintained after decellularization. Decellularized ECM from both protocols was used as a substrate or as soluble factor, in both cases showing good cell attachment and growth capabilities. Interestingly, the 1-day protocol was capable of promoting greater differentiation than the 4-day protocol, probably due to its capacity to remove a similar amount of cell nuclei, while better conserving the biochemical and structural components of the cerebral ECM. Still, some limitations of this study include the need to evaluate the response in other biologically relevant cell types, as well as a more detailed characterization of the components in the decellularized ECM of the different brain subregions. In conclusion, our results show differences in neuronal maturation depending on the region of the brain used to produce the scaffolds. Complex organs such as the brain have subregions with very different initial cellular and biochemical conditions that should be considered for decellularization to minimize exposure to immunogenic components, while retaining bioactive factors conducive to regeneration. [Figure: see text] Impact statement The present study offers new knowledge about the production of decellularized extracellular matrix scaffolds from specific regions of the porcine brain, with a direct comparison of their effect on in vitro neuronal maturation. Our results show differences in neuronal maturation depending on the region of the brain used to produce the scaffolds, suggesting that it is necessary to consider the initial cellular content of the source tissue and its bioactive capacity for the production of an effective regenerative therapy for stroke.
Collapse
Affiliation(s)
- Diego Reginensi
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama.,School of Medicine, Universidad de Panamá, Panama, Republic of Panama.,Biomedical Engineering Program, Universidad Latina de Panamá, Panama, Republic of Panama
| | - Didio Ortiz
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Andrea Pravia
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama.,Biotechnology Program, Universidad Latina de Panamá, Panama, Republic of Panama
| | - Andrea Burillo
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Félix Morales
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Carly Morgan
- CREO-MIHRT Program, University of California, Santa Cruz, California.,Materials Science & Engineering Program, University of Washington, Seattle, Washington
| | - Lindsay Jimenez
- CREO-MIHRT Program, University of California, Santa Cruz, California.,Biomedical Engineering Program, University of Connecticut, Storrs, Connecticut
| | - Kunjan R Dave
- The Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Neurology Department, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Miguel A Perez-Pinzon
- The Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Neurology Department, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Rolando A Gittens
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama.,Centro de Biodiversidad y Descubrimiento de Drogas, INDICASAT AIP, City of Knowledge, Panama, Republic of Panama
| |
Collapse
|
9
|
Krishnaswamy VR, Benbenishty A, Blinder P, Sagi I. Demystifying the extracellular matrix and its proteolytic remodeling in the brain: structural and functional insights. Cell Mol Life Sci 2019; 76:3229-3248. [PMID: 31197404 PMCID: PMC11105229 DOI: 10.1007/s00018-019-03182-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022]
Abstract
The extracellular matrix (ECM) plays diverse roles in several physiological and pathological conditions. In the brain, the ECM is unique both in its composition and in functions. Furthermore, almost all the cells in the central nervous system contribute to different aspects of this intricate structure. Brain ECM, enriched with proteoglycans and other small proteins, aggregate into distinct structures around neurons and oligodendrocytes. These special structures have cardinal functions in the normal functioning of the brain, such as learning, memory, and synapse regulation. In this review, we have compiled the current knowledge about the structure and function of important ECM molecules in the brain and their proteolytic remodeling by matrix metalloproteinases and other enzymes, highlighting the special structures they form. In particular, the proteoglycans in brain ECM, which are essential for several vital functions, are emphasized in detail.
Collapse
Affiliation(s)
| | - Amit Benbenishty
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Pablo Blinder
- Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv, Israel
- Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
10
|
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: 6.0] [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]
|
11
|
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: 13.2] [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
|
12
|
Nieuwenhuis B, Haenzi B, Andrews MR, Verhaagen J, Fawcett JW. Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 2018; 93:1339-1362. [PMID: 29446228 PMCID: PMC6055631 DOI: 10.1111/brv.12398] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/23/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Integrins are cell surface receptors that form the link between extracellular matrix molecules of the cell environment and internal cell signalling and the cytoskeleton. They are involved in several processes, e.g. adhesion and migration during development and repair. This review focuses on the role of integrins in axonal regeneration. Integrins participate in spontaneous axonal regeneration in the peripheral nervous system through binding to various ligands that either inhibit or enhance their activation and signalling. Integrin biology is more complex in the central nervous system. Integrins receptors are transported into growing axons during development, but selective polarised transport of integrins limits the regenerative response in adult neurons. Manipulation of integrins and related molecules to control their activation state and localisation within axons is a promising route towards stimulating effective regeneration in the central nervous system.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
| | | | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
- Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam1081 HVAmsterdamThe Netherlands
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Centre of Reconstructive NeuroscienceInstitute of Experimental Medicine142 20Prague 4Czech Republic
| |
Collapse
|
13
|
The Tenascin-C-Derived Peptide VSWRAPTA Promotes Neuronal Branching Via Transcellular Activation of the Focal Adhesion Kinase (FAK) and the ERK1/2 Signaling Pathway In Vitro. Mol Neurobiol 2018; 56:632-647. [DOI: 10.1007/s12035-018-1108-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/03/2018] [Indexed: 12/27/2022]
|
14
|
Motalleb R, Berns EJ, Patel P, Gold J, Stupp SI, Kuhn HG. In vivo migration of endogenous brain progenitor cells guided by an injectable peptide amphiphile biomaterial. J Tissue Eng Regen Med 2018; 12:e2123-e2133. [PMID: 29327429 DOI: 10.1002/term.2644] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 11/22/2017] [Accepted: 01/02/2018] [Indexed: 12/28/2022]
Abstract
Biomaterials hold great promise in helping the adult brain regenerate and rebuild after trauma. Peptide amphiphiles (PAs) are highly versatile biomaterials, gelling and forming macromolecular structures when exposed to physiological levels of electrolytes. We are here reporting on the first ever in vivo use of self-assembling PA carrying a Tenascin-C signal (E2 Ten-C PA) for the redirection of endogenous neuroblasts in the rodent brain. The PA forms highly aligned nanofibers, displaying the migratory sequence of Tenascin-C glycoprotein as epitope. In this in vivo work, we have formed in situ a gel of aligned PA nanofibers presenting a migratory Tenascin-C signal sequence in the ventral horn of the rostral migratory stream, creating a track reaching the neocortex. Seven days posttransplant, doublecortin positive cells were observed migrating inside and alongside the injected biomaterial, reaching the cortex. We observed a 24-fold increase in number of redirected neuroblasts for the E2 Ten-C PA-injected animals compared to control. We also found injecting the E2 Ten-C PA to cause minimal neuroinflammatory response. Analysing GFAP+ astrocytes and Iba1+ microglia activation, the PA does not elicit a stronger neuroinflammatory response than would be expected from a small needle stab wound. Redirecting endogenous neuroblasts and increasing the number of cells reaching a site of injury using PAs may open up new avenues for utilizing the pool of neuroblasts and neural stem cells within the adult brain for regenerating damaged brain tissue and replacing neurons lost to injury.
Collapse
Affiliation(s)
- Reza Motalleb
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Eric J Berns
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Piyush Patel
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Julie Gold
- Department of Applied Physics, Biological Physics, Chalmers University of Technology, Gothenburg, Sweden
| | - Samuel I Stupp
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.,Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.,Department of Chemistry, Northwestern University, Evanston, IL, USA.,Department of Medicine, Northwestern University, Chicago, IL, USA.,Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | - H Georg Kuhn
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Charité - Universitätsmedizin Berlin, Neurocure Cluster of Excellence, Berlin, Germany
| |
Collapse
|
15
|
Reinhard J, Roll L, Faissner A. Tenascins in Retinal and Optic Nerve Neurodegeneration. Front Integr Neurosci 2017; 11:30. [PMID: 29109681 PMCID: PMC5660115 DOI: 10.3389/fnint.2017.00030] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/03/2017] [Indexed: 02/04/2023] Open
Abstract
Tenascins represent key constituents of the extracellular matrix (ECM) with major impact on central nervous system (CNS) development. In this regard, several studies indicate that they play a crucial role in axonal growth and guidance, synaptogenesis and boundary formation. These functions are not only important during development, but also for regeneration under several pathological conditions. Additionally, tenascin-C (Tnc) represents a key modulator of the immune system and inflammatory processes. In the present review article, we focus on the function of Tnc and tenascin-R (Tnr) in the diseased CNS, specifically after retinal and optic nerve damage and degeneration. We summarize the current view on both tenascins in diseases such as glaucoma, retinal ischemia, age-related macular degeneration (AMD) or diabetic retinopathy. In this context, we discuss their expression profile, possible functional relevance, remodeling of the interacting matrisome and tenascin receptors, especially under pathological conditions.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
16
|
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: 6.7] [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
|
17
|
Role of Matricellular Proteins in Disorders of the Central Nervous System. Neurochem Res 2016; 42:858-875. [DOI: 10.1007/s11064-016-2088-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022]
|
18
|
Faissner A. REVIEW ■ : Glial Derived Extracellular Matrix Components: Important Roles in Axon Growth and Guidance. Neuroscientist 2016. [DOI: 10.1177/107385849700300610] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Axon growth and guidance, and the correct recognition of distant targets by growth cones rank among the most spectacular achievements of the developing nervous system. The establishment and reformation of adequate networks and the plasticity of synaptic connections are vital for the function and the restoration of the nervous system under conditions of health and disease. Therefore, considerable efforts have been devoted to the elucidation of the molecular and cellular bases of the establishment of interneuronal con nections. It is well established that interactions between neurons and astrocytes are of regulatory importance in this context. Thus, astroglia guides migrating neurons and advancing growth cones to their destination. On the other hand, astrocytes design transient boundaries that deflect axons and segregate groups of neurons, and form scars involved in the inhibition of axonal regeneration after lesion. This duplicity of astroglia is presumably mediated by various gene families. Among these, extracellular matrix (ECM) con stituents seem particularly suited to embody and mediate the ambivalence of astrocytes because these compounds appear to exert either conducive or inhibitory/repulsive effects depending on interacting cell types and conditions. Furthermore, ECM constituents are upregulated by astrocytes upon lesion and con tribute to the construction of glial scars. This review focuses on this class of compounds and their possible functions in the wiring of neural networks. NEUROSCIENTIST 3:371-380, 1997
Collapse
|
19
|
Astrocyte scar formation aids central nervous system axon regeneration. Nature 2016; 532:195-200. [PMID: 27027288 DOI: 10.1038/nature17623] [Citation(s) in RCA: 1227] [Impact Index Per Article: 153.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/26/2016] [Indexed: 12/20/2022]
Abstract
Transected axons fail to regrow in the mature central nervous system. Astrocytic scars are widely regarded as causal in this failure. Here, using three genetically targeted loss-of-function manipulations in adult mice, we show that preventing astrocyte scar formation, attenuating scar-forming astrocytes, or ablating chronic astrocytic scars all failed to result in spontaneous regrowth of transected corticospinal, sensory or serotonergic axons through severe spinal cord injury (SCI) lesions. By contrast, sustained local delivery via hydrogel depots of required axon-specific growth factors not present in SCI lesions, plus growth-activating priming injuries, stimulated robust, laminin-dependent sensory axon regrowth past scar-forming astrocytes and inhibitory molecules in SCI lesions. Preventing astrocytic scar formation significantly reduced this stimulated axon regrowth. RNA sequencing revealed that astrocytes and non-astrocyte cells in SCI lesions express multiple axon-growth-supporting molecules. Our findings show that contrary to the prevailing dogma, astrocyte scar formation aids rather than prevents central nervous system axon regeneration.
Collapse
|
20
|
Kwiatkowska M, Reinhard J, Roll L, Kraft N, Dazert S, Faissner A, Volkenstein S. The expression pattern and inhibitory influence of Tenascin-C on the growth of spiral ganglion neurons suggest a regulatory role as boundary formation molecule in the postnatal mouse inner ear. Neuroscience 2016; 319:46-58. [PMID: 26812032 DOI: 10.1016/j.neuroscience.2016.01.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/08/2015] [Accepted: 01/15/2016] [Indexed: 02/02/2023]
Abstract
Sensorineural hearing loss, as a consequence of acoustic trauma, aging, genetic defects or ototoxic drugs, is highly associated with irreversible damage of cochlear hair cells (HCs) and secondary degeneration of spiral ganglion (SG) cells. Cochlear implants (CIs), which bypass the lost HC function by direct electrical stimulation of the remaining auditory neurons, offer an effective therapy option. Several studies imply that components of the extracellular matrix (ECM) have a great impact on the adhesion and growth of spiral ganglion neurons (SGNs) during development. Based on these findings, ECM proteins might act as bioactive CI substrates to optimize the electrode-nerve interface and to improve efficacy of these implants. In the present study, we focused on the ECM glycoproteins Tenascin-C (TN-C), Laminin (LN), and Fibronectin (FN), which show a prominent expression along the growth route of SGNs and the niche around HCs during murine postnatal development in vivo. We compared their influence on adhesion, neurite length, and neurite number of SGNs in vitro. Moreover, we studied the expression of the chondroitin sulfate proteoglycan (CSPG) dermatan sulfate-dependent proteoglycan-1 (DSD-1-PG), an interaction partner of TN-C. In sum, our in vitro data suggest that TN-C acts as an anti-adhesive and inhibitory factor for the growth of SGNs. The DSD-1 carbohydrate epitope is specifically localized to HC stereocilia and SG fibers. Interestingly, TN-C and the DSD-1-PG exhibit a mutually exclusive expression pattern, with the exception of a very restricted region beneath the habenula perforata, where SG neurites grow through the basilar membrane (BM) toward the HCs. The complementary expression of TN-C, LN, FN, and the DSD-1 epitope suggests that TN-C may act as an important boundary formation molecule in the developing postnatal mouse inner ear, which makes it a promising candidate to regulate neurite outgrowth in the light of CIs.
Collapse
Affiliation(s)
- M Kwiatkowska
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - J Reinhard
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - L Roll
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - N Kraft
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - S Dazert
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - A Faissner
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - S Volkenstein
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany.
| |
Collapse
|
21
|
Abstract
Tenascin-C is a large, multimodular, extracellular matrix glycoprotein that exhibits a very restricted pattern of expression but an enormously diverse range of functions. Here, we discuss the importance of deciphering the expression pattern of, and effects mediated by, different forms of this molecule in order to fully understand tenascin-C biology. We focus on both post transcriptional and post translational events such as splicing, glycosylation, assembly into a 3D matrix and proteolytic cleavage, highlighting how these modifications are key to defining tenascin-C function.
Collapse
Key Words
- AD1/AD2, additional domain 1/ additional domain 2
- ADAMTS, a disintegrin and metalloproteinase with thrombospondin motifs
- ASMCs, aortic smooth muscle cells
- BDNF, brain derived neurotrophic factor
- BHKs, baby hamster kidney cells
- BMP, bone morphogenetic protein
- CA19–9, carbohydrate antigen 19–9
- CALEB, chicken acidic leucine-rich EGF-like domain containing brain protein
- CEA, carcinoembryonic antigen
- CNS, central nervous system
- CRC, colorectal carcinomas
- CTGF, connective tissue growth factor
- DCIS, ductal carcinoma in-situ
- ECM, extracellular matrix
- EDA-FN, extra domain A containing fibronectin
- EDB-FN, extra domain B containing fibronectin
- EGF-L, epidermal growth factor-like
- EGF-R, epidermal growth factor receptor
- ELISPOT, enzyme-linked immunospot assay
- FBG, fibrinogen-like globe
- FGF2, fibroblast growth factor 2
- FGF4, fibroblast growth factor 4
- FN, fibronectin
- FNIII, fibronectin type III-like repeat
- GMEM, glioma-mesenchymal extracellular matrix antigen
- GPI, glycosylphosphatidylinositol
- HB-EGF, heparin-binding EGF-like growth factor
- HCEs, immortalized human corneal epithelial cell line
- HGF, hepatocyte growth factor
- HNK-1, human natural killer-1
- HSPGs, heparan sulfate proteoglycans
- HUVECs, human umbilical vein endothelial cells
- ICC, immunocytochemistry
- IF, immunofluorescence
- IFNγ, interferon gamma
- IGF, insulin-like growth factor
- IGF-BP, insulin-like growth factor-binding protein
- IHC, immunohistochemistry
- IL, interleukin
- ISH, in situ hybridization
- LPS, lipopolysaccharide
- MMP, matrix metalloproteinase
- MPNSTs, malignant peripheral nerve sheath tumors
- Mr, molecular mass
- NB, northern blot
- NF-kB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NK, natural killer cells
- NSCLC, non-small cell lung carcinoma
- NSCs, neural stem cells
- NT, neurotrophin
- PAMPs, pathogen-associated molecular patterns
- PDGF, platelet derived growth factor
- PDGF-Rβ, platelet derived growth factor receptor β
- PIGF, phosphatidylinositol-glycan biosynthesis class F protein
- PLCγ, phospholipase-C gamma
- PNS, peripheral nervous system
- PTPRζ1, receptor-type tyrosine-protein phosphatase zeta
- RA, rheumatoid arthritis
- RCC, renal cell carcinoma
- RD, rhabdomyosarcoma
- RGD, arginylglycylaspartic acid
- RT-PCR, real-time polymerase chain reaction
- SB, Southern blot
- SCC, squamous cell carcinoma
- SMCs, smooth muscle cells
- SVZ, sub-ventricular zone
- TA, tenascin assembly domain
- TGFβ, transforming growth factor β
- TIMP, tissue inhibitor of metalloproteinases
- TLR4, toll-like receptor 4
- TNFα, tumor necrosis factor α
- TSS, transcription start site
- UBC, urothelial bladder cancer
- UCC, urothelial cell carcinoma
- VEGF, vascular endothelial growth factor
- VSMCs, vascular smooth muscle cells
- VZ, ventricular zone
- WB, immunoblot/ western blot
- bFGF, basic fibroblast growth factor
- biosynthesis
- c, charged
- cancer
- ccRCC, clear cell renal cell carcinoma
- chRCC, chromophobe-primary renal cell carcinoma
- development
- glycosylation
- mAb, monoclonal antibody
- matrix assembly
- mitogen-activated protein kinase, MAPK
- pHo, extracellular pH
- pRCC, papillary renal cell carcinoma
- proteolytic cleavage
- siRNA, small interfering RNA
- splicing
- tenascin-C
- therapeutics
- transcription
Collapse
Affiliation(s)
- Sean P Giblin
- a Nuffield Department of Orthopaedics; Rheumatology and Musculoskeletal Sciences ; Kennedy Institute of Rheumatology; University of Oxford ; Oxford , UK
| | | |
Collapse
|
22
|
Schira J, Falkenberg H, Hendricks M, Waldera-Lupa DM, Kögler G, Meyer HE, Müller HW, Stühler K. Characterization of Regenerative Phenotype of Unrestricted Somatic Stem Cells (USSC) from Human Umbilical Cord Blood (hUCB) by Functional Secretome Analysis. Mol Cell Proteomics 2015; 14:2630-43. [PMID: 26183719 DOI: 10.1074/mcp.m115.049312] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Indexed: 12/13/2022] Open
Abstract
Stem cell transplantation is a promising therapeutic strategy to enhance axonal regeneration after spinal cord injury. Unrestricted somatic stem cells (USSC) isolated from human umbilical cord blood is an attractive stem cell population available at GMP grade without any ethical concerns. It has been shown that USSC transplantation into acute injured rat spinal cords leads to axonal regrowth and significant locomotor recovery, yet lacking cell replacement. Instead, USSC secrete trophic factors enhancing neurite growth of primary cortical neurons in vitro. Here, we applied a functional secretome approach characterizing proteins secreted by USSC for the first time and validated candidate neurite growth promoting factors using primary cortical neurons in vitro. By mass spectrometric analysis and exhaustive bioinformatic interrogation we identified 1156 proteins representing the secretome of USSC. Using Gene Ontology we revealed that USSC secretome contains proteins involved in a number of relevant biological processes of nerve regeneration such as cell adhesion, cell motion, blood vessel formation, cytoskeleton organization and extracellular matrix organization. We found for instance that 31 well-known neurite growth promoting factors like, e.g. neuronal growth regulator 1, NDNF, SPARC, and PEDF span the whole abundance range of USSC secretome. By the means of primary cortical neurons in vitro assays we verified SPARC and PEDF as significantly involved in USSC mediated neurite growth and therewith underline their role in improved locomotor recovery after transplantation. From our data we are convinced that USSC are a valuable tool in regenerative medicine as USSC's secretome contains a comprehensive network of trophic factors supporting nerve regeneration not only by a single process but also maintained its regenerative phenotype by a multitude of relevant biological processes.
Collapse
Affiliation(s)
- Jessica Schira
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany; §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Heiner Falkenberg
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Marion Hendricks
- §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Daniel M Waldera-Lupa
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Gesine Kögler
- ¶Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Medical Center, Düsseldorf, Germany
| | - Helmut E Meyer
- ‖Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund, Germany
| | - Hans Werner Müller
- §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; **Biologisch-Medizinisches Forschungszentrum (BMFZ), Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Kai Stühler
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany; **Biologisch-Medizinisches Forschungszentrum (BMFZ), Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| |
Collapse
|
23
|
Brösicke N, Sallouh M, Prior LM, Job A, Weberskirch R, Faissner A. Extracellular Matrix Glycoprotein-Derived Synthetic Peptides Differentially Modulate Glioma and Sarcoma Cell Migration. Cell Mol Neurobiol 2015; 35:741-53. [PMID: 25783630 DOI: 10.1007/s10571-015-0170-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 03/04/2015] [Indexed: 12/20/2022]
Abstract
Glycoproteins of the extracellular matrix (ECM) regulate proliferation, migration, and differentiation in numerous cell lineages. ECM functions are initiated by small peptide sequences embedded in large constituents that are recognized by specific cellular receptors. In this study, we have investigated the biological effects of peptides derived from collagen type IV and tenascin-C compared to the well-known RGD peptide originally discovered in fibronectin. The influence of glycoproteins and corresponding peptides on the migration of the glioma cell lines U-251-MG and U-373-MG and the sarcoma line S-117 was studied. When the cell lines were tested in a modified Boyden chamber assay on filters coated with the ECM glycoproteins, glioma cells showed a strong migration response on tenascin-C and the basal lamina constituent collagen IV, in contrast to S-117 cells. In order to identify relevant stimulatory motifs, peptides derived from fibronectin (6NHX-GRGDSF), tenascin-C (TN-C, VSWRAPTA), and collagen type IV (MNYYSNS) were compared, either applied in solution in combination with ECM glycoprotein substrates, in solution in the presence of untreated membranes, or coated on the filters of the Boyden chambers. Using this strategy, we could identify the novel tenascin-C-derived peptide motif VSWRAPTA as a migration stimulus for glioma cells. Furthermore, while kin peptides generally blocked the effects of the respective homologous ECM proteins, unexpected effects were observed in heterologous situations. There, in several cases, addition of soluble peptides strongly boosted the response to the coated ECM proteins. We propose that peptides may synergize or antagonize each other by stimulating different signaling pathways.
Collapse
Affiliation(s)
- Nicole Brösicke
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Reinhard J, Joachim SC, Faissner A. Extracellular matrix remodeling during retinal development. Exp Eye Res 2015; 133:132-40. [DOI: 10.1016/j.exer.2014.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 10/25/2022]
|
25
|
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: 23] [Impact Index Per Article: 2.6] [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
|
26
|
Irnidayanti Y. Existence of vimentin and GFAP protein expressions as a result of 2-Methoxyethanol administration in cerebral cortex tissue of Swiss Webster mice (Mus musculus): an immunohistochemical analysis. Pak J Biol Sci 2014; 17:876-883. [PMID: 26035935 DOI: 10.3923/pjbs.2014.876.883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Une of the plastic-based materials widely used in the plastics industry in various countries is ester phthalate. This compound will be oxidized in the body into 2-methoxyethanol (2-ME). The effect of 2-ME on human health and environment depends on the number, duration and the frequency of exposure. Recently, the incidence of brain damage tends to increase. In the last decade, it has been widely reported the negative effects of chemical pollutants to the environment. The aim of this study were to know the existence of the expression of Vimentin and GFAP proteins caused by 2-ME on the histological structure of the cerebral cortex of mice fetal during the prenatal period on gestation day 14 (GD 14) and day 18 (GD 18). The 2-ME compound was injected intraperitoneally with a dose of 7.5 mmol kg(-1) of body weight at GD-10. The result showed that there is a change in existence Vimentin protein in the cerebral cortex fetal of treated mice at GD 14, which is very conspicuous. Meanwhile, a change in existence of GFAP protein in cerebral cortex fetal of treated mice at GD 14, have relatively no difference from controls and no impact on histological structure changes of the cerebral corteks at GD 14. The change in existence of Vimentin protein in the cerebral cortex fetal of treated mice at GD 14 have an impact on histological structure of the cerebral cortex of mice treated at GD 18. It is believed that the impact is due to the effects of 2-methoxyethanol.
Collapse
|
27
|
Schreiber J, Schachner M, Schumacher U, Lorke DE. Extracellular matrix alterations, accelerated leukocyte infiltration and enhanced axonal sprouting after spinal cord hemisection in tenascin-C-deficient mice. Acta Histochem 2013; 115:865-78. [PMID: 23701962 DOI: 10.1016/j.acthis.2013.04.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 01/08/2023]
Abstract
The extracellular matrix glycoprotein tenascin-C has been implicated in wound repair and axonal growth. Its role in mammalian spinal cord injury is largely unknown. In vitro it can be both neurite-outgrowth promoting and repellent. To assess its effects on glial reactions, extracellular matrix formation, and axonal regrowth/sprouting in vivo, 20 tenascin-C-deficient and 20 wild type control mice underwent lumbar spinal cord hemisection. One, three, seven and fourteen days post-surgery, cryostat sections of the spinal cord were examined by conventional histology and by immunohistochemistry using antibodies against F4/80 (microglia/macrophage), GFAP (astroglia), neurofilament, fibronectin, laminin and collagen type IV. Fibronectin immunoreactivity was significantly down-regulated in tenascin-C-deficient mice. Moreover, fourteen days after injury, immunodensity of neurofilament-positive fibers was two orders of magnitude higher along the incision edges of tenascin-C-deficient mice as compared to control mice. In addition, lymphocyte infiltration was seen two days earlier in tenascin-C-deficient mice than in control mice and neutrophil infiltration was increased seven days after injury. The increase in thin neurofilament positive fibers in tenascin-C-deficient mice indicates that lack of tenascin-C alters the inflammatory reaction and extracellular matrix composition in a way that penetration of axonal fibers into spinal cord scar tissue may be facilitated.
Collapse
Affiliation(s)
- Jenny Schreiber
- University Medical Center Hamburg-Eppendorf, Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, Martinistraße 52, 20246 Hamburg, Germany
| | | | | | | |
Collapse
|
28
|
Brösicke N, van Landeghem FKH, Scheffler B, Faissner A. Tenascin-C is expressed by human glioma in vivo and shows a strong association with tumor blood vessels. Cell Tissue Res 2013; 354:409-30. [DOI: 10.1007/s00441-013-1704-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 07/12/2013] [Indexed: 12/17/2022]
|
29
|
Volpato FZ, Führmann T, Migliaresi C, Hutmacher DW, Dalton PD. Using extracellular matrix for regenerative medicine in the spinal cord. Biomaterials 2013; 34:4945-55. [PMID: 23597407 DOI: 10.1016/j.biomaterials.2013.03.057] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/20/2013] [Indexed: 12/12/2022]
Abstract
Regeneration within the mammalian central nervous system (CNS) is limited, and traumatic injury often leads to permanent functional motor and sensory loss. The lack of regeneration following spinal cord injury (SCI) is mainly caused by the presence of glial scarring, cystic cavitation and a hostile environment to axonal growth at the lesion site. The more prominent experimental treatment strategies focus mainly on drug and cell therapies, however recent interest in biomaterial-based strategies are increasing in number and breadth. Outside the spinal cord, approaches that utilize the extracellular matrix (ECM) to promote tissue repair show tremendous potential for various application including vascular, skin, bone, cartilage, liver, lung, heart and peripheral nerve tissue engineering (TE). Experimentally, it is unknown if these approaches can be successfully translated to the CNS, either alone or in combination with synthetic biomaterial scaffolds. In this review we outline the first attempts to apply the potential of ECM-based biomaterials and combining cell-derived ECM with synthetic scaffolds.
Collapse
Affiliation(s)
- Fabio Zomer Volpato
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove 4059, Australia
| | | | | | | | | |
Collapse
|
30
|
Staphylococcal superantigen-like protein 8 (SSL8) binds to tenascin C and inhibits tenascin C-fibronectin interaction and cell motility of keratinocytes. Biochem Biophys Res Commun 2013; 433:127-32. [PMID: 23485472 DOI: 10.1016/j.bbrc.2013.02.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 02/08/2013] [Indexed: 01/25/2023]
Abstract
Staphylococcal superantigen-like protein (SSL), a family of exotoxins composed of 14 SSLs, exhibits no superantigenic activity despite of its structural similarity with superantigens. Several SSLs have been revealed to bind to host immune molecules such as IgA, IgG, complement and cell surface molecules expressed on immune cells, but the physiological function of SSL family has not been fully identified. In this study we attempted to isolate host target proteins of SSLs from human breast milk using SSLs-conjugated Sepharose. SSL8-conjugated Sepharose specifically recovered tenascin C (TNC), a multimodular and multifunctional extracellular matrix protein. Pull down analysis using SSL8-conjugated Sepharose and recombinant truncated fragments of TNC revealed that SSL8 interacts with fibronectin (FN) type III repeats 1-5 of TNC. The interaction of TNC with immobilized FN was attenuated, the scratch wound closure by HaCaT human keratinocytes was delayed and the inhibition of cell spreading on FN by TNC was recovered in the presence of SSL8. These findings suggest that SSL8 binds to TNC, thereby inhibits the TNC-FN interaction and motility of keratinocytes. The present study added a novel role of SSL family protein as an interrupting molecule against the function of extracellular matrix.
Collapse
|
31
|
Jakovcevski I, Miljkovic D, Schachner M, Andjus PR. Tenascins and inflammation in disorders of the nervous system. Amino Acids 2012; 44:1115-27. [DOI: 10.1007/s00726-012-1446-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 12/10/2012] [Indexed: 12/20/2022]
|
32
|
Wiese S, Karus M, Faissner A. Astrocytes as a source for extracellular matrix molecules and cytokines. Front Pharmacol 2012; 3:120. [PMID: 22740833 PMCID: PMC3382726 DOI: 10.3389/fphar.2012.00120] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 06/06/2012] [Indexed: 12/19/2022] Open
Abstract
Research of the past 25 years has shown that astrocytes do more than participating and building up the blood-brain barrier and detoxify the active synapse by reuptake of neurotransmitters and ions. Indeed, astrocytes express neurotransmitter receptors and, as a consequence, respond to stimuli. Within the tripartite synapse, the astrocytes owe more and more importance. Besides the functional aspects the differentiation of astrocytes has gained a more intensive focus. Deeper knowledge of the differentiation processes during development of the central nervous system might help explaining and even help treating neurological diseases like Alzheimer’s disease, Amyotrophic lateral sclerosis, Parkinsons disease, and psychiatric disorders in which astrocytes have been shown to play a role. Specific differentiation of neural stem cells toward the astroglial lineage is performed as a multi-step process. Astrocytes and oligodendrocytes develop from a multipotent stem cell that prior to this has produced primarily neuronal precursor cells. This switch toward the more astroglial differentiation is regulated by a change in receptor composition on the cell surface and responsiveness to Fibroblast growth factor and Epidermal growth factor (EGF). The glial precursor cell is driven into the astroglial direction by signaling molecules like Ciliary neurotrophic factor, Bone Morphogenetic Proteins, and EGF. However, the early astrocytes influence their environment not only by releasing and responding to diverse soluble factors but also express a wide range of extracellular matrix (ECM) molecules, in particular proteoglycans of the lectican family and tenascins. Lately these ECM molecules have been shown to participate in glial development. In this regard, especially the matrix protein Tenascin C (Tnc) proved to be an important regulator of astrocyte precursor cell proliferation and migration during spinal cord development. Nevertheless, ECM molecules expressed by reactive astrocytes are also known to act mostly in an inhibitory fashion under pathophysiological conditions. Thus, we further summarize resent data concerning the role of chondroitin sulfate proteoglycans and Tnc under pathological conditions.
Collapse
Affiliation(s)
- Stefan Wiese
- Group for Molecular Cell Biology, Department for Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany
| | | | | |
Collapse
|
33
|
Roll L, Mittmann T, Eysel UT, Faissner A. The laser lesion of the mouse visual cortex as a model to study neural extracellular matrix remodeling during degeneration, regeneration and plasticity of the CNS. Cell Tissue Res 2012; 349:133-45. [DOI: 10.1007/s00441-011-1313-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/20/2011] [Indexed: 02/06/2023]
|
34
|
Myers JP, Santiago-Medina M, Gomez TM. Regulation of axonal outgrowth and pathfinding by integrin-ECM interactions. Dev Neurobiol 2011; 71:901-23. [PMID: 21714101 PMCID: PMC3192254 DOI: 10.1002/dneu.20931] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Developing neurons use a combination of guidance cues to assemble a functional neural network. A variety of proteins immobilized within the extracellular matrix (ECM) provide specific binding sites for integrin receptors on neurons. Integrin receptors on growth cones associate with a number of cytosolic adaptor and signaling proteins that regulate cytoskeletal dynamics and cell adhesion. Recent evidence suggests that soluble growth factors and classic axon guidance cues may direct axon pathfinding by controlling integrin-based adhesion. Moreover, because classic axon guidance cues themselves are immobilized within the ECM and integrins modulate cellular responses to many axon guidance cues, interactions between activated receptors modulate cell signals and adhesion. Ultimately, growth cones control axon outgrowth and pathfinding behaviors by integrating distinct biochemical signals to promote the proper assembly of the nervous system. In this review, we discuss our current understanding how ECM proteins and their associated integrin receptors control neural network formation.
Collapse
Affiliation(s)
- Jonathan P Myers
- Department of Neuroscience, Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
35
|
Dobbertin A, Czvitkovich S, Theocharidis U, Garwood J, Andrews MR, Properzi F, Lin R, Fawcett JW, Faissner A. Analysis of combinatorial variability reveals selective accumulation of the fibronectin type III domains B and D of tenascin-C in injured brain. Exp Neurol 2010; 225:60-73. [PMID: 20451518 DOI: 10.1016/j.expneurol.2010.04.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 04/23/2010] [Accepted: 04/30/2010] [Indexed: 10/19/2022]
Abstract
Tenascin-C (Tnc) is a multimodular extracellular matrix glycoprotein that is markedly upregulated in CNS injuries where it is primarily secreted by reactive astrocytes. Different Tnc isoforms can be generated by the insertion of variable combinations of one to seven (in rats) alternatively spliced distinct fibronectin type III (FnIII) domains to the smallest variant. Each spliced FnIII repeat mediates specific actions on neurite outgrowth, neuron migration or adhesion. Hence, different Tnc isoforms might differentially influence CNS repair. We explored the expression pattern of Tnc variants after cortical lesions and after treatment of astrocytes with various cytokines. Using RT-PCR, we observed a strong upregulation of Tnc transcripts containing the spliced FnIII domains B or D in injured tissue at 2-4 days post-lesion (dpl). Looking at specific combinations, we showed a dramatic increase of Tnc isoforms harboring the neurite outgrowth-promoting BD repeat with both the B and D domains being adjacent to each other. Isoforms containing only the axon growth-stimulating spliced domain D were also dramatically enhanced after injury. Injury-induced increase of Tnc proteins comprising the domain D was confirmed by Western Blotting and immunostaining of cortical lesions. In contrast, the FnIII modules C and AD1 were weakly modulated after injury. The growth cone repulsive A1A2A4 domains were poorly expressed in normal and injured tissue but the smallest isoform, which is also repellant, was highly expressed after injury. Expression of the shortest Tnc isoform and of variants containing B, D or BD, was strongly upregulated in cultured astrocytes after TGFbeta1 treatment, suggesting that TGFbeta1 could mediate, at least in part, the injury-induced upregulation of these isoforms. We identified complex injury-induced differential regulations of Tnc isoforms that may well influence axonal regeneration and repair processes in the damaged CNS.
Collapse
Affiliation(s)
- Alexandre Dobbertin
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University of Bochum, 44780 Bochum, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Comparative screening of glial cell types reveals extracellular matrix that inhibits retinal axon growth in a chondroitinase ABC-resistant fashion. Glia 2009; 57:1420-38. [DOI: 10.1002/glia.20860] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
37
|
DNA vaccination against neurite growth inhibitors to enhance functional recovery following traumatic brain injury. ACTA NEUROCHIRURGICA. SUPPLEMENT 2009. [PMID: 19388343 DOI: 10.1007/978-3-211-85578-2_66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
BACKGROUND Myelin-associated proteins contribute to failure of axon regeneration in the injured central nervous system of the adult. METHODS In this study, we employed a recombinant DNA vaccine encoding the myelin-derived inhibitors NogoA, myelin-associated glycoprotein (MAG) and tenascin-R (TnR), so as to effect the production of antibodies against these myelin-related antigens, in a rodent head injury model and ascertained its potential for promoting axonal plasticity and functional recovery. Adult rats underwent lateral fluid percussion at the left sensorimotor cortex (SMC) and treatment with the DNA vaccine before or after injury. Behavioral tests and neuroanatomical tract tracing was carried out. FINDINGS The vaccinated rats showed improved corticorubral plasticity and functional recovery compared to control groups. CONCLUSIONS This suggests that a DNA vaccination approach may provide a promising strategy for promoting repair after traumatic brain injury.
Collapse
|
38
|
The glia-derived extracellular matrix glycoprotein tenascin-C promotes embryonic and postnatal retina axon outgrowth via the alternatively spliced fibronectin type III domain TNfnD. ACTA ACUST UNITED AC 2009; 4:271-83. [DOI: 10.1017/s1740925x09990020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Tenascin-C (Tnc) is an astrocytic multifunctional extracellular matrix (ECM) glycoprotein that potentially promotes or inhibits neurite outgrowth. To investigate its possible functions for retinal development, explants from embryonic day 18 (E18) rat retinas were cultivated on culture substrates composed of poly-d-lysine (PDL), or PDL additionally coated with Tnc or laminin (LN)-1, which significantly increased fiber length. When combined with LN, Tnc induced axon fasciculation that reduced the apparent number of outgrowing fibers. In order to circumscribe the stimulatory region, Tnc-derived fibronectin type III (TNfn) domains fused to the human Ig-Fc-fragment TNfnD6-Fc, TNfnBD-Fc, TNFnA1A2-Fc and TNfnA1D-Fc were studied. The fusion proteins TNfnBD-Fc and to a lesser degree TNfnA1D-Fc were stimulatory when compared with the Ig-Fc-fragment protein without insert. In contrast, the combination TNfnA1A2-Fc reduced fiber outgrowth beneath the values obtained for the Ig-Fc domain, indicating potential inhibitory properties. The monoclonal J1/tn2 antibody (clone 578) that is specific for domain TNfnD blocked the stimulatory properties of the TNfn-Fc fusions. When postnatal day 7 retinal ganglion cells were used rather that explants, Tnc and Tnc-derived proteins proved permissive for neurite outgrowth. The present study highlights a strong retinal axon growth-promoting activity of the Tnc domain TNfnD, which is modulated by neighboring domains.
Collapse
|
39
|
Gervasi NM, Kwok JC, Fawcett JW. Role of extracellular factors in axon regeneration in the CNS: implications for therapy. Regen Med 2009; 3:907-23. [PMID: 18947312 DOI: 10.2217/17460751.3.6.907] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The glial scar that forms after an injury to the CNS contains molecules that are inhibitory to axon growth. Understanding of the mechanisms of inhibition has allowed the development of therapeutic strategies aimed at promoting axon regeneration. Promising results have been obtained in animal models, and some therapies are undergoing clinical trials. This offers great hope for achievement of functional recovery after CNS injury.
Collapse
Affiliation(s)
- Noreen M Gervasi
- Cambridge University Centre for Brain Repair, ED Adrian Building, Forvie Site, Robinson Way, Cambridge CB22PY, UK.
| | | | | |
Collapse
|
40
|
Royland JE, Parker JS, Gilbert ME. A genomic analysis of subclinical hypothyroidism in hippocampus and neocortex of the developing rat brain. J Neuroendocrinol 2008; 20:1319-38. [PMID: 19094080 DOI: 10.1111/j.1365-2826.2008.01793.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Hypothyroidism during pregnancy and the early postnatal period has severe neurological consequences for the developing offspring. The impact of milder degrees of perturbation of the thyroid axis as encompassed in conditions of subclinical hypothyroidism and hypothyroxinemia, however, has not been established. The present investigation examined the effects of graded levels of hypothyroidism, from subclinical to severe, on global gene expression in the developing rodent brain. Thyroid hormone insufficiency was induced by administration of propylthiouracil (PTU) to pregnant rats via drinking water from gestational day 6 until sacrifice of pups prior to weaning. In the first study a specialised microarray, the Affymetrix Rat Neurobiology array RN_U34, was used to contrast gene expression in the hippocampus of animals exposed to 0 or 10 ppm (10 mg/l) PTU, a treatment producing severe hypothyroidism. In the second study, a more complete genome array (Affymetrix Rat 230A) was used to compare gene expression in the neocortex and hippocampus of postnatal day (PN) 14 animals experiencing graded degrees of thyroid hormone insufficiency induced by delivery of 0, 1, 2 or 3 ppm PTU to the dam. Dose-dependent up- and down-regulation were observed for gene transcripts known to play critical roles in brain development and brain function. Expression levels of a subset of approximately 25 genes in each brain region were altered at a dose of PTU (1 ppm) that induced mild hypothyroxinemia in dams and pups. These data indicate that genes driving important developmental processes are sensitive to relatively modest perturbations of the thyroid axis, and that the level of gene expression is related to the degree of hormone reduction. Altered patterns of gene expression during critical windows of brain development indicate that thyroid disease must be viewed as a continuum and that conditions typically considered 'subclinical' may induce structural and functional abnormalities in the developing central nervous system.
Collapse
Affiliation(s)
- J E Royland
- Neurotoxicology Division, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | | | | |
Collapse
|
41
|
Meiners S, Ahmed I, Ponery AS, Amor N, Harris SL, Ayres V, Fan Y, Chen Q, Delgado-Rivera R, Babu AN. Engineering electrospun nanofibrillar surfaces for spinal cord repair: a discussion. POLYM INT 2007. [DOI: 10.1002/pi.2383] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
42
|
Tochigi M, Zhang X, Ohashi J, Hibino H, Otowa T, Rogers M, Kato T, Okazaki Y, Kato N, Tokunaga K, Sasaki T. Association study between the TNXB locus and schizophrenia in a Japanese population. Am J Med Genet B Neuropsychiatr Genet 2007; 144B:305-9. [PMID: 17192952 DOI: 10.1002/ajmg.b.30441] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The chromosome 6p21-24 region, which contains the human leukocyte antigen (HLA) region, has been suggested as an important locus for a susceptibility gene for schizophrenia. Recently, a significant association between schizophrenia and the TNXB locus, located immediately telomeric of the NOTCH4 locus in the HLA region, was observed. Few studies have further investigated the region in schizophrenia. In the present study, we investigated the region in a Japanese population. Subjects included 241 patients with schizophrenia and 290 controls. Twenty-six single nucleotide polymorphisms (SNPs) and the corresponding haplotypes were analyzed. As a result, exactly the same SNPs in the TNXB locus (rs1009382 and rs204887) as in the previous study were associated with schizophrenia (P = 0.034 and 0.034, respectively, uncorrected). A SNP (rs2071287) in the NOTCH4 locus and haplotype around it were also suggested to associate with the disease, consistent with another previous study (P = 0.041 and permutation P = 0.024, respectively, uncorrected). Although these associations became insignificant after Bonferroni correction, the findings might provide support for the association of the TNXB locus or its adjacent region of the NOTCH4 locus with schizophrenia.
Collapse
Affiliation(s)
- Mamoru Tochigi
- Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Bunkyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Faissner A, Heck N, Dobbertin A, Garwood J. DSD-1-Proteoglycan/Phosphacan and Receptor Protein Tyrosine Phosphatase-Beta Isoforms during Development and Regeneration of Neural Tissues. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:25-53. [PMID: 16955703 DOI: 10.1007/0-387-30128-3_3] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Interactions between neurons and glial cells play important roles in regulating key events of development and regeneration of the CNS. Thus, migrating neurons are partly guided by radial glia to their target, and glial scaffolds direct the growth and directional choice of advancing axons, e.g., at the midline. In the adult, reactive astrocytes and myelin components play a pivotal role in the inhibition of regeneration. The past years have shown that astrocytic functions are mediated on the molecular level by extracellular matrix components, which include various glycoproteins and proteoglycans. One important, developmentally regulated chondroitin sulfate proteoglycan is DSD-1-PG/phosphacan, a glial derived proteoglycan which represents a splice variant of the receptor protein tyrosine phosphatase (RPTP)-beta (also known as PTP-zeta). Current evidence suggests that this proteoglycan influences axon growth in development and regeneration, displaying inhibitory or stimulatory effects dependent on the mode of presentation, and the neuronal lineage. These effects seem to be mediated by neuronal receptors of the Ig-CAM superfamily.
Collapse
Affiliation(s)
- Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University, Bochum, Germany
| | | | | | | |
Collapse
|
44
|
Abstract
Neural recognition molecules were discovered and characterized initially for their functional roles in cell adhesion as regulators of affinity between cells and the extracellular matrix in vitro. They were then recognized as mediators or co-receptors which trigger signal transduction mechanisms affecting cell adhesion and de-adhesion. Their involvement in contact attraction and repulsion relies on cell-intrinsic properties that are modulated by the spatial contexts of their expression at particular stages of ontogenetic development, in synaptic plasticity and during regeneration after injury. The functional roles of recognition molecules in cell proliferation and migration, determination of developmental fate, growth cone guidance, and synapse formation, stabilization and modulation have been well documented not only by in vitro, but also by in vivo studies that have been greatly aided by generation of genetically altered mice. More recently, the functions of recognition molecules have been investigated under conditions of neural repair and manipulated using a broad range of genetic and pharmacological approaches to achieve a beneficial outcome. The principal aim of most therapeutically oriented approaches has been to neutralize inhibitory factors. However, less attention has been paid to enhancing repair by stimulating the stimulatory factors. When considering potential therapeutic strategies, it is worth considering that a single recognition molecule can possess domains that are conducive or repellent and that the spatial distribution of recognition molecules can determine the overall function: Recognition molecules may be repellent for neurite outgrowth when presented as barriers or steep-concentration gradients and conducive when presented as uniform substrates. The focus of this review will be on the more recent attempts to study the conducive mechanisms with the expectation that they may be able to tip the balance from a regeneration inhospitable to a hospitable environment. It is likely that a combination of the two principles, as multifactorial as each principle may be in itself, will be of therapeutic value in humans.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Universität Hamburg, Hamburg, Germany
| | | |
Collapse
|
45
|
Metzger M, Bartsch S, Bartsch U, Bock J, Schachner M, Braun K. Regional and cellular distribution of the extracellular matrix protein tenascin-C in the chick forebrain and its role in neonatal learning. Neuroscience 2006; 141:1709-19. [PMID: 16797128 DOI: 10.1016/j.neuroscience.2006.05.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Revised: 05/08/2006] [Accepted: 05/09/2006] [Indexed: 11/22/2022]
Abstract
The juvenile brain's pronounced synaptic plasticity in response to early experience and learning events is related to the fact that the genetically pre-programmed molecular machinery mediating neuronal development and synapse formation, is activated throughout postnatal brain development and thereby can be recruited for learning and long-term memory formation. In situ hybridization and immunocytochemistry experiments revealed that tenascin-C, one candidate molecule which we suspect to be involved in neonatal learning, is expressed in the forebrain of domestic chicks around the sensitive period during which auditory filial imprinting takes place. The involvement of tenascin-C in this juvenile learning task was tested by injections of monoclonal antibodies directed to distinct domains of the tenascin-C molecule into the avian prefrontal cortex analog, the medio-rostral nidopallium/mesopallium (formerly termed medio-rostral neostriatum/hyperstriatum ventrale), a forebrain area which has been shown to be critically involved in auditory filial imprinting. Injections of monoclonal antibody Tn 68, which is directed against a cell-binding domain of the tenascin-C molecule, strongly reduced the imprinting rate, as opposed to injections of the monoclonal antibody Tn 578, which binds to a domain involved in neurite outgrowth. Double labeling immunohistochemistry revealed that tenascin-C is associated with neurons which express the Ca(2+)-binding protein parvalbumin, and displays a staining pattern highly reminiscent of perineuronal nets of the extracellular matrix. These results indicate that a distinct domain of tenascin-C is functionally involved in the juvenile learning process of filial imprinting and further suggest a critical role of a specific neuronal subpopulation.
Collapse
Affiliation(s)
- M Metzger
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 1524, São Paulo, SP 05508-900, Brazil
| | | | | | | | | | | |
Collapse
|
46
|
Pruss T, Kranz EU, Niere M, Volkmer H. A regulated switch of chick neurofascin isoforms modulates ligand recognition and neurite extension. Mol Cell Neurosci 2006; 31:354-65. [PMID: 16314110 DOI: 10.1016/j.mcn.2005.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 09/30/2005] [Accepted: 10/12/2005] [Indexed: 10/25/2022] Open
Abstract
Neural cell adhesion molecule neurofascin regulates the induction of neurite outgrowth, the establishment of synaptic connectivity and myelination. Neurofascin isoforms are generated by spatially and temporally controlled alternative splicing. Isoform NF166 is predominantly expressed in dorsal root ganglia from embryonal day 5 (E5) to E8, and a further neurofascin isoform NF185 appears at E9. Expression of neurofascin and its binding partner axonin-1 on sensory fibers implies functional interactions for neurite outgrowth. E7 sensory neurons require NF166-axonin-1 interactions for neurite extension, accordingly. The contribution of NF166-axonin-1 interaction for neurite outgrowth decreases in parallel with the appearance of NF185 on sensory neurons at E9. This finding may be explained by (1) alleviated intrinsic capability to use axonin-1 as a cellular receptor and (2) reduced binding of axonin-1 to NF185. Finally, NF166, but not NF185, serves as a cellular receptor for neurite induction via homophilic interactions with a neurofascin substrate.
Collapse
Affiliation(s)
- Thomas Pruss
- NMI Naturwissenschaftliches und Medizinisches Institut an der Universität Tübingen, NMI, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | | | | | | |
Collapse
|
47
|
Radim J, Dubovy P. Immunohistochemical labelling of components of the endoneurial extracellular matrix of intact and rhizotomized dorsal and ventral spinal roots of the rat--a quantitative evaluation using image analysis. Acta Histochem 2006; 107:453-62. [PMID: 16414104 DOI: 10.1016/j.acthis.2005.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Revised: 10/26/2005] [Accepted: 10/31/2005] [Indexed: 12/14/2022]
Abstract
The endoneurial extracellular matrix (ECM) molecules are involved in cell signalling during nervous system development and regeneration. Quantitative differences of immunofluorescence labelling for chondroitin sulfate proteoglycan (CSPG), fibronectin (FN), tenascin-C (TN-C), and thrombospondin (TSP) were evaluated in intact rat dorsal and ventral roots and dorsal and ventral roots 2 and 4 weeks after rhizotomy using image analysis. The distal stumps of spinal roots displayed increased immunolabelling for the molecules with higher immunofluorescence in dorsal than in ventral roots up to 2 weeks from transection. Four weeks after rhizotomy, the immunoreactivity for CSPG, TN-C and TSP decreased in dorsal and increased in ventral root stumps, although a higher level of immunofluorescence for FN remained in both dorsal and ventral root stumps 4 weeks after injury in comparison to 2 weeks after injury. We suggest that the amount of some ECM molecules changed differentially 2 and 4 weeks after rhizotomy to create an appropriate environment in the endoneurium for early and later regrowth of sensory and motor axons. The results presented here are the first report of differences between the endoneurial ECM content of damaged afferent and motor nerve fibers. In addition, the immunohistochemical detection of individual ECM molecules indicated that final extrinsic conditions stimulating the regrowth of regenerating axons probably arise from a balance of both growth-promoting and -inhibiting molecules in the endoneurium.
Collapse
Affiliation(s)
- Jancalek Radim
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Kamenice 3, CZ-625 00 Brno, Czech Republic
| | | |
Collapse
|
48
|
Liu HY, Nur-E-Kamal A, Schachner M, Meiners S. Neurite guidance by the FnC repeat of human tenascin-C: neurite attraction vs. neurite retention. Eur J Neurosci 2005; 22:1863-72. [PMID: 16262626 DOI: 10.1111/j.1460-9568.2005.04383.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The alternatively spliced fibronectin type-III repeat C of human tenascin-C (fnC) provides directional cues to elongating neurites in vitro. When given a choice at an interface with poly L-lysine (PLL), rat cerebellar granule neurites preferentially crossed onto fnC (defined herein as neurite attraction) whereas neurites originating on fnC preferentially remained on fnC (defined as neurite retention). Guidance motifs were further refined using synthetic peptides spanning the sequence of fnC. We found that a peptide with amino acid sequence DINPYGFTVSWMASE was sufficient to attract and retain neurites. Peptides with alterations in NPYG facilitated neurite retention but not attraction and, conversely, molecules with alterations in ASE facilitated neurite attraction but not retention. Hence neurite attraction and neurite retention mediated by fnC are separable events that can be independently regulated. This property may prove valuable for the strategic design of peptide reagents for use in strategies to facilitate directed axonal regrowth following CNS injury.
Collapse
Affiliation(s)
- Hsing-Yin Liu
- Department of Pharmacology, UMDNJ-Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
49
|
Garwood J, Garcion E, Dobbertin A, Heck N, Calco V, ffrench-Constant C, Faissner A. The extracellular matrix glycoprotein Tenascin-C is expressed by oligodendrocyte precursor cells and required for the regulation of maturation rate, survival and responsiveness to platelet-derived growth factor. Eur J Neurosci 2005; 20:2524-40. [PMID: 15548197 DOI: 10.1111/j.1460-9568.2004.03727.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Analysis of Tenascin-C (TN-C) knockout mice revealed novel roles for this extracellular matrix (ECM) protein in regulation of the developmental programme of oligodendrocyte precursor cells (OPCs), their maturation into myelinating oligodendrocytes and sensitivity to growth factors. A major component of the ECM of developing nervous tissue, TN-C was expressed in zones of proliferation, migration and morphogenesis. Examination of TN-C knockout mice showed roles for TN-C in control of OPC proliferation and migration towards zones of myelination [E. Garcion et al. (2001) Development, 128, 2485-2496]. Extending our studies of TN-C effects on OPC development we found that OPCs can endogenously express TN-C protein. This expression covered the whole range of possible TN-C isoforms and could be strongly up-regulated by leukaemia inhibitory factor and ciliary neurotrophic factor, cytokines known to modulate OPC proliferation and survival. Comparative analysis of TN-C knockout OPCs with wild-type OPCs reveals an accelerated rate of maturation in the absence of TN-C, with earlier morphological differentiation and precocious expression of myelin basic protein. TN-C knockout OPCs plated on poly-lysine displayed higher levels of apoptosis than wild-type OPCs and there was also an earlier loss of responsiveness to the protective effects of platelet-derived growth factor (PDGF), indicating that TN-C has anti-apoptotic effects that may be associated with PDGF signalling. The existence of mechanisms to compensate for the absence of TN-C in the knockout is indicated by the development of oligodendrocytes derived from TN-C knockout neurospheres. These were present in equivalent proportions to those found in wild-type neurospheres but displayed enhanced myelin membrane formation.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens/metabolism
- Blotting, Western/methods
- Brain/cytology
- Brain/embryology
- Brain/growth & development
- Brain/metabolism
- Bromodeoxyuridine/metabolism
- Cell Count/methods
- Cell Differentiation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Cerebral Cortex/cytology
- Chondroitin Sulfates/metabolism
- Cytokines/pharmacology
- Embryo, Mammalian
- Gene Expression Regulation, Developmental/drug effects
- Humans
- Immunohistochemistry/methods
- In Situ Hybridization/methods
- In Situ Nick-End Labeling/methods
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Biological
- Myelin Basic Protein/metabolism
- Nerve Tissue Proteins/metabolism
- Oligodendroglia/drug effects
- Oligodendroglia/metabolism
- Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology
- Platelet-Derived Growth Factor/pharmacology
- Protein Tyrosine Phosphatases/metabolism
- Proteoglycans/metabolism
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Receptor-Like Protein Tyrosine Phosphatases, Class 5
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Stem Cells/drug effects
- Stem Cells/metabolism
- Tenascin/genetics
- Tenascin/physiology
- Time Factors
Collapse
Affiliation(s)
- Jeremy Garwood
- LNDR, CNRS 5, rue Blaise Pascal, 67084 Strasbourg Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
50
|
Heck N, Garwood J, Loeffler JP, Larmet Y, Faissner A. Differential upregulation of extracellular matrix molecules associated with the appearance of granule cell dispersion and mossy fiber sprouting during epileptogenesis in a murine model of temporal lobe epilepsy. Neuroscience 2005; 129:309-24. [PMID: 15501589 DOI: 10.1016/j.neuroscience.2004.06.078] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2004] [Indexed: 01/06/2023]
Abstract
We have investigated changes in the extracellular matrix of the hippocampus associated with the early progression of epileptogenesis in a murine model of temporal lobe epilepsy using immunohistochemistry. In the first week following intrahippocampal injection of the glutamate agonist, domoate, there is a latent period at the end of which begins a sequential upregulation of extracellular matrix (ECM) molecules in the granule cell layer of the dentate gyrus, beginning with neurocan and tenascin-C. This expression precedes the characteristic dispersion of the granule cell layer which is evident at 14 days post-injection when the first recurrent seizures can be recorded. At this stage, an upregulation of the chondroitin sulfate proteoglycan, phosphacan, the DSD-1 chondroitin sulfate motif, and the HNK-1 oligosaccharide are also observed. The expression of these molecules is localized differentially in the epileptogenic dentate gyrus, especially in the sprouting molecular layer, where a strong upregulation of phosphacan, tenascin-C, and HNK-1 is observed but there is no expression of the proteoglycan, neurocan, nor of the DSD-1 chondroitin sulfate motif. Hence, it appears that granule cell layer dispersion is accompanied by a general increase in the ECM, while mossy fiber sprouting in the molecular layer is associated with a more restricted repertoire. In contrast to these changes, the expression of the ECM glycoproteins, laminin and fibronectin, both of which are frequently implicated in tissue remodelling events, showed no changes associated with either granule cell dispersion or mossy fiber sprouting, indicating that the epileptogenic plasticity of the hippocampus is accompanied by ECM interactions that are characteristic of the CNS.
Collapse
Affiliation(s)
- N Heck
- LNDR, Centre de Neurochimie du CNRS, 5, rue Blaise Pascal, 67084 Strasbourg, France
| | | | | | | | | |
Collapse
|