1
|
Hilton BJ, Griffin JM, Fawcett JW, Bradke F. Neuronal maturation and axon regeneration: unfixing circuitry to enable repair. Nat Rev Neurosci 2024; 25:649-667. [PMID: 39164450 DOI: 10.1038/s41583-024-00849-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Mammalian neurons lose the ability to regenerate their central nervous system axons as they mature during embryonic or early postnatal development. Neuronal maturation requires a transformation from a situation in which neuronal components grow and assemble to one in which these components are fixed and involved in the machinery for effective information transmission and computation. To regenerate after injury, neurons need to overcome this fixed state to reactivate their growth programme. A variety of intracellular processes involved in initiating or sustaining neuronal maturation, including the regulation of gene expression, cytoskeletal restructuring and shifts in intracellular trafficking, have been shown to prevent axon regeneration. Understanding these processes will contribute to the identification of targets to promote repair after injury or disease.
Collapse
Affiliation(s)
- Brett J Hilton
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Jarred M Griffin
- Laboratory for Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - James W Fawcett
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK.
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine Czech Academy of Science (CAS), Prague, Czechia.
| | - Frank Bradke
- Laboratory for Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
2
|
Wang XW, Yang SG, Hu MW, Wang RY, Zhang C, Kosanam AR, Ochuba AJ, Jiang JJ, Luo X, Guan Y, Qian J, Liu CM, Zhou FQ. Histone methyltransferase Ezh2 coordinates mammalian axon regeneration via regulation of key regenerative pathways. J Clin Invest 2024; 134:e163145. [PMID: 38015636 PMCID: PMC10849760 DOI: 10.1172/jci163145] [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: 06/29/2022] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
Current treatments for neurodegenerative diseases and neural injuries face major challenges, primarily due to the diminished regenerative capacity of neurons in the mammalian CNS as they mature. Here, we investigated the role of Ezh2, a histone methyltransferase, in regulating mammalian axon regeneration. We found that Ezh2 declined in the mouse nervous system during maturation but was upregulated in adult dorsal root ganglion neurons following peripheral nerve injury to facilitate spontaneous axon regeneration. In addition, overexpression of Ezh2 in retinal ganglion cells in the CNS promoted optic nerve regeneration via both histone methylation-dependent and -independent mechanisms. Further investigation revealed that Ezh2 fostered axon regeneration by orchestrating the transcriptional silencing of genes governing synaptic function and those inhibiting axon regeneration, while concurrently activating various factors that support axon regeneration. Notably, we demonstrated that GABA transporter 2, encoded by Slc6a13, acted downstream of Ezh2 to control axon regeneration. Overall, our study underscores the potential of modulating chromatin accessibility as a promising strategy for promoting CNS axon regeneration.
Collapse
Affiliation(s)
- Xue-Wei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Shu-Guang Yang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Rui-Ying Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chi Zhang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anish R. Kosanam
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Arinze J. Ochuba
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jing-Jing Jiang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Perez-Gianmarco L, Kukley M. Understanding the Role of the Glial Scar through the Depletion of Glial Cells after Spinal Cord Injury. Cells 2023; 12:1842. [PMID: 37508505 PMCID: PMC10377788 DOI: 10.3390/cells12141842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Spinal cord injury (SCI) is a condition that affects between 8.8 and 246 people in a million and, unlike many other neurological disorders, it affects mostly young people, causing deficits in sensory, motor, and autonomic functions. Promoting the regrowth of axons is one of the most important goals for the neurological recovery of patients after SCI, but it is also one of the most challenging goals. A key event after SCI is the formation of a glial scar around the lesion core, mainly comprised of astrocytes, NG2+-glia, and microglia. Traditionally, the glial scar has been regarded as detrimental to recovery because it may act as a physical barrier to axon regrowth and release various inhibitory factors. However, more and more evidence now suggests that the glial scar is beneficial for the surrounding spared tissue after SCI. Here, we review experimental studies that used genetic and pharmacological approaches to ablate specific populations of glial cells in rodent models of SCI in order to understand their functional role. The studies showed that ablation of either astrocytes, NG2+-glia, or microglia might result in disorganization of the glial scar, increased inflammation, extended tissue degeneration, and impaired recovery after SCI. Hence, glial cells and glial scars appear as important beneficial players after SCI.
Collapse
Affiliation(s)
- Lucila Perez-Gianmarco
- Achucarro Basque Center for Neuroscience, 48940 Leioa, PC, Spain
- Department of Neurosciences, University of the Basque Country, 48940 Leioa, PC, Spain
| | - Maria Kukley
- Achucarro Basque Center for Neuroscience, 48940 Leioa, PC, Spain
- IKERBASQUE Basque Foundation for Science, 48009 Bilbao, PC, Spain
| |
Collapse
|
4
|
The Effect of Different Routes of Xenogeneic Mesenchymal Stem Cell Transplantation on the Regenerative Potential of Spinal Cord Injury. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00290-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
5
|
Gu J, Gao B, Zafar H, Chu B, Feng X, Ni Y, Xu L, Bao R. Thermo-sensitive hydrogel combined with SHH expressed RMSCs for rat spinal cord regeneration. Front Bioeng Biotechnol 2022; 10:1001396. [PMID: 36338109 PMCID: PMC9634076 DOI: 10.3389/fbioe.2022.1001396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Purpose: Spinal cord injury (SCI) has a damaging impact on patients, amid being a worldwide problem with no effective treatment. Herein, we reported a method for functional therapy of SCI in rats, wherein we combined thermo-sensitive hydrogel with Sonic Hedgehog (SHH) expressed in rat bone-marrow derived mesenchymal stem cells (RMSCs). Methods: Bone marrow-derived mesenchymal stem cells (BMSCs) were isolated from Sprague-Dawley (SD) female rats. The SHH was optimized and transferred into RMSCs via cationic liposomes, while thermo-sensitive hydrogel was reformed with hyaluronate (HA) and Pluronic F127. Then, a rat model with SCI was established accordingly by male SD rats and randomized into sham, model, RMSCs with hydrogel and SHH-RMSCs with hydrogel. The evaluation of SCI repair based on Basso, Beattie Bresnahanlocomotor rating scale (BBB scale) and inclined plate score. Immunofluorescence, immunohistochemistry and hematoxylin-eosin were utilized to explore the expression of protein (GFAP, GAP43, NF200 and MBP) and histopathology. Results: It was demonstrated that transfection of SHH with cationic liposomes exhibited more effect in RMSCs than lipofectamine 2000. As shown in SEM, 3.5% HA-F127 demonstrated porous structure. In the MTT and dead/live assay, 3.5% HA-F127 showed good biocompatibility for RMSCs. Both RMSCs and SHH-RMSCs groups could significantly promote BBB and inclined plate scores (p < 0.01) compared with the model. Furthermore, the SHH-RMSC group was significantly improved than RMSC with the expression of related proteins, where NF200, MBP, and GAP43 were principally enhanced with the GFAP expression being virtually down-regulated. Conclusion: All in all, the results suggested that transplantation of RMSCs with SHH could improve the function of SCI and promote nerve regeneration.
Collapse
Affiliation(s)
- Jun Gu
- School of Medicine, Yangzhou University, Yangzhou, China
- Department of Orthopedics, Xishan People’s Hospital, Wuxi, China
- *Correspondence: Jun Gu, ; Hajra Zafar,
| | - Biao Gao
- School of Medicine, Yangzhou University, Yangzhou, China
- Wuxi Xishan District Ehu Town Health Center, Wuxi, China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jun Gu, ; Hajra Zafar,
| | - Bo Chu
- Department of Orthopedics, Xishan People’s Hospital, Wuxi, China
| | - Xiaojun Feng
- Department of Orthopedics, Xishan People’s Hospital, Wuxi, China
| | - Yinjie Ni
- Department of Orthopedics, Xishan People’s Hospital, Wuxi, China
| | - Lin Xu
- Department of Orthopedics, Xishan People’s Hospital, Wuxi, China
| | - Rui Bao
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, China
| |
Collapse
|
6
|
Tang F, Brune JE, Chang MY, Reeves SR, Altemeier WA, Frevert CW. Defining the versican interactome in lung health and disease. Am J Physiol Cell Physiol 2022; 323:C249-C276. [PMID: 35649251 PMCID: PMC9291419 DOI: 10.1152/ajpcell.00162.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022]
Abstract
The extracellular matrix (ECM) imparts critical mechanical and biochemical information to cells in the lungs. Proteoglycans are essential constituents of the ECM and play a crucial role in controlling numerous biological processes, including regulating cellular phenotype and function. Versican, a chondroitin sulfate proteoglycan required for embryonic development, is almost absent from mature, healthy lungs and is reexpressed and accumulates in acute and chronic lung disease. Studies using genetically engineered mice show that the versican-enriched matrix can be pro- or anti-inflammatory depending on the cellular source or disease process studied. The mechanisms whereby versican develops a contextual ECM remain largely unknown. The primary goal of this review is to provide an overview of the interaction of versican with its many binding partners, the "versican interactome," and how through these interactions, versican is an integrator of complex extracellular information. Hopefully, the information provided in this review will be used to develop future studies to determine how versican and its binding partners can develop contextual ECMs that control select biological processes. Although this review focuses on versican and the lungs, what is described can be extended to other proteoglycans, tissues, and organs.
Collapse
Affiliation(s)
- Fengying Tang
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Jourdan E Brune
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Mary Y Chang
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Stephen R Reeves
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington
| | - William A Altemeier
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Charles W Frevert
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
7
|
The Extracellular Matrix Proteins Tenascin-C and Tenascin-R Retard Oligodendrocyte Precursor Maturation and Myelin Regeneration in a Cuprizone-Induced Long-Term Demyelination Animal Model. Cells 2022; 11:cells11111773. [PMID: 35681468 PMCID: PMC9179356 DOI: 10.3390/cells11111773] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
Oligodendrocytes are the myelinating cells of the central nervous system. The physiological importance of oligodendrocytes is highlighted by diseases such as multiple sclerosis, in which the myelin sheaths are degraded and the axonal signal transmission is compromised. In a healthy brain, spontaneous remyelination is rare, and newly formed myelin sheaths are thinner and shorter than the former ones. The myelination process requires the migration, proliferation, and differentiation of oligodendrocyte precursor cells (OPCs) and is influenced by proteins of the extracellular matrix (ECM), which consists of a network of glycoproteins and proteoglycans. In particular, the glycoprotein tenascin-C (Tnc) has an inhibitory effect on the differentiation of OPCs and the remyelination efficiency of oligodendrocytes. The structurally similar tenascin-R (Tnr) exerts an inhibitory influence on the formation of myelin membranes in vitro. When Tnc knockout oligodendrocytes were applied to an in vitro myelination assay using artificial fibers, a higher number of sheaths per single cell were obtained compared to the wild-type control. This effect was enhanced by adding brain-derived neurotrophic factor (BDNF) to the culture system. Tnr−/− oligodendrocytes behaved differently in that the number of formed sheaths per single cell was decreased, indicating that Tnr supports the differentiation of OPCs. In order to study the functions of tenascin proteins in vivo Tnc−/− and Tnr−/− mice were exposed to Cuprizone-induced demyelination for a period of 10 weeks. Both Tnc−/− and Tnr−/− mouse knockout lines displayed a significant increase in the regenerating myelin sheath thickness after Cuprizone treatment. Furthermore, in the absence of either tenascin, the number of OPCs was increased. These results suggest that the fine-tuning of myelin regeneration is regulated by the major tenascin proteins of the CNS.
Collapse
|
8
|
A Monoclonal Anti-HMGB1 Antibody Attenuates Neurodegeneration in an Experimental Animal Model of Glaucoma. Int J Mol Sci 2022; 23:ijms23084107. [PMID: 35456925 PMCID: PMC9028318 DOI: 10.3390/ijms23084107] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is a crucial process for the loss of retinal ganglion cells (RGC), a major characteristic of glaucoma. High expression of high-mobility group box protein 1 (HMGB1) plays a detrimental role in inflammatory processes and is elevated in the retinas of glaucoma patients. Therefore, this study aimed to investigate the effects of the intravitreal injection of an anti-HMGB1 monoclonal antibody (anti-HMGB1 Ab) in an experimental animal model of glaucoma. Two groups of Spraque Dawley rats received episcleral vein occlusion to chronically elevate intraocular pressure (IOP): (1) the IgG group, intravitreal injection of an unspecific IgG as a control, n = 5, and (2) the HMGB1 group, intravitreal injection of an anti-HMGB1 Ab, n = 6. IOP, retinal nerve fiber layer thickness (RNFLT), and the retinal flash response were monitored longitudinally. Post-mortem examinations included immunohistochemistry, microarray, and mass spectrometric analysis. RNFLT was significantly increased in the HMGB1 group compared with the IgG group (p < 0.001). RGC density showed improved neuronal cell survival in the retina in HMGB1 compared with the IgG group (p < 0.01). Mass spectrometric proteomic analysis of retinal tissue showed an increased abundance of RNA metabolism-associated heterogeneous nuclear ribonucleoproteins (hnRNPs), such as hnRNP U, D, and H2, in animals injected with the anti-HMGB1 Ab, indicating that the application of the antibody may cause increased gene expression. Microarray analysis showed a significantly decreased expression of C-X-C motif chemokine ligand 8 (CXCL8, p < 0.05) and connective tissue growth factor (CTGF, p < 0.01) in the HMGB1 group. Thus, these data suggest that intravitreal injection of anti-HMGB1 Ab reduced HMGB1-dependent inflammatory signaling and mediated RGC neuroprotection.
Collapse
|
9
|
Bauch J, Ort SV, Ulc A, Faissner A. Tenascins Interfere With Remyelination in an Ex Vivo Cerebellar Explant Model of Demyelination. Front Cell Dev Biol 2022; 10:819967. [PMID: 35372366 PMCID: PMC8965512 DOI: 10.3389/fcell.2022.819967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/24/2022] [Indexed: 01/02/2023] Open
Abstract
Oligodendrocytes form myelin membranes and thereby secure the insulation of axons and the rapid conduction of action potentials. Diseases such as multiple sclerosis highlight the importance of this glial cell population for brain function. In the adult brain, efficient remyelination following the damage to oligodendrocytes is compromised. Myelination is characterized by proliferation, migration, and proper integration of oligodendrocyte precursor cells (OPCs). These processes are among others controlled by proteins of the extracellular matrix (ECM). As a prominent representative ECM molecule, tenascin-C (Tnc) exerts an inhibitory effect on the migration and differentiation of OPCs. The structurally similar paralogue tenascin-R (Tnr) is known to promote the differentiation of oligodendrocytes. The model of lysolecithin-induced demyelination of cerebellar slice cultures represents an important tool for the analysis of the remyelination process. Ex vivo cerebellar explant cultures of Tnc−/− and Tnr−/− mouse lines displayed enhanced remyelination by forming thicker myelin membranes upon exposure to lysolecithin. The inhibitory effect of tenascins on remyelination could be confirmed when demyelinated wildtype control cultures were exposed to purified Tnc or Tnr protein. In that approach, the remyelination efficiency decreased in a dose-dependent manner with increasing concentrations of ECM molecules added. In order to examine potential roles in a complex in vivo environment, we successfully established cuprizone-based acute demyelination to analyze the remyelination behavior after cuprizone withdrawal in SV129, Tnc−/−, and Tnr−/− mice. In addition, we documented by immunohistochemistry in the cuprizone model the expression of chondroitin sulfate proteoglycans that are inhibitory for the differentiation of OPCs. In conclusion, inhibitory properties of Tnc and Tnr for myelin membrane formation could be demonstrated by using an ex vivo approach.
Collapse
|
10
|
Zhang Q, Li Y, Zhuo Y. Synaptic or Non-synaptic? Different Intercellular Interactions with Retinal Ganglion Cells in Optic Nerve Regeneration. Mol Neurobiol 2022; 59:3052-3072. [PMID: 35266115 PMCID: PMC9016027 DOI: 10.1007/s12035-022-02781-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
Axons of adult neurons in the mammalian central nervous system generally fail to regenerate by themselves, and few if any therapeutic options exist to reverse this situation. Due to a weak intrinsic potential for axon growth and the presence of strong extrinsic inhibitors, retinal ganglion cells (RGCs) cannot regenerate their axons spontaneously after optic nerve injury and eventually undergo apoptosis, resulting in permanent visual dysfunction. Regarding the extracellular environment, research to date has generally focused on glial cells and inflammatory cells, while few studies have discussed the potentially significant role of interneurons that make direct connections with RGCs as part of the complex retinal circuitry. In this study, we provide a novel angle to summarize these extracellular influences following optic nerve injury as "intercellular interactions" with RGCs and classify these interactions as synaptic and non-synaptic. By discussing current knowledge of non-synaptic (glial cells and inflammatory cells) and synaptic (mostly amacrine cells and bipolar cells) interactions, we hope to accentuate the previously neglected but significant effects of pre-synaptic interneurons and bring unique insights into future pursuit of optic nerve regeneration and visual function recovery.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
11
|
Echternacht SR, Chacon MA, Leckenby JI. Central versus peripheral nervous system regeneration: is there an exception for cranial nerves? Regen Med 2021; 16:567-579. [PMID: 34075805 DOI: 10.2217/rme-2020-0096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
There exists a dichotomy in regenerative capacity between the PNS and CNS, which poses the question - where do cranial nerves fall? Through the discussion of the various cells and processes involved in axonal regeneration, we will evaluate whether the assumption that cranial nerve regeneration is analogous to peripheral nerve regeneration is valid. It is evident from this review that much remains to be clarified regarding both PNS and CNS regeneration. Furthermore, it is not clear if cranial nerves follow the PNS model, CNS model or possess an alternative novel regenerative process altogether. Future research should continue to focus on elucidating how cranial nerves regenerate; and the various cellular interactions, molecules and pathways involved.
Collapse
Affiliation(s)
- Scott R Echternacht
- University of Rochester School of Medicine & Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.,Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| | - Miranda A Chacon
- Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA.,Department of Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| | - Jonathan I Leckenby
- Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| |
Collapse
|
12
|
Extracellular Matrix Remodeling in the Retina and Optic Nerve of a Novel Glaucoma Mouse Model. BIOLOGY 2021; 10:biology10030169. [PMID: 33668263 PMCID: PMC7996343 DOI: 10.3390/biology10030169] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/14/2021] [Accepted: 02/16/2021] [Indexed: 12/23/2022]
Abstract
Simple Summary Glaucoma is a leading cause of blindness worldwide, and increased age and intraocular pressure (IOP) are the major risk factors. Glaucoma is characterized by the death of nerve cells and the loss of optic nerve fibers. Recently, evidence has accumulated indicating that proteins in the environment of nerve cells, called the extracellular matrix (ECM), play an important role in glaucomatous neurodegeneration. Depending on its constitution, the ECM can influence either the survival or the death of nerve cells. Thus, the aim of our study was to comparatively explore alterations of various ECM molecules in the retina and optic nerve of aged control and glaucomatous mice with chronic IOP elevation. Interestingly, we observed elevated levels of blood vessel and glial cell-associated ECM components in the glaucomatous retina and optic nerve, which could be responsible for various pathological processes. A better understanding of the underlying signaling mechanisms may help to develop new diagnostic and therapeutic strategies for glaucoma patients. Abstract Glaucoma is a neurodegenerative disease that is characterized by the loss of retinal ganglion cells (RGC) and optic nerve fibers. Increased age and intraocular pressure (IOP) elevation are the main risk factors for developing glaucoma. Mice that are heterozygous (HET) for the mega-karyocyte protein tyrosine phosphatase 2 (PTP-Meg2) show chronic and progressive IOP elevation, severe RGCs loss, and optic nerve damage, and represent a valuable model for IOP-dependent primary open-angle glaucoma (POAG). Previously, evidence accumulated suggesting that glaucomatous neurodegeneration is associated with the extensive remodeling of extracellular matrix (ECM) molecules. Unfortunately, little is known about the exact ECM changes in the glaucomatous retina and optic nerve. Hence, the goal of the present study was to comparatively explore ECM alterations in glaucomatous PTP-Meg2 HET and control wild type (WT) mice. Due to their potential relevance in glaucomatous neurodegeneration, we specifically analyzed the expression pattern of the ECM glycoproteins fibronectin, laminin, tenascin-C, and tenascin-R as well as the proteoglycans aggrecan, brevican, and members of the receptor protein tyrosine phosphatase beta/zeta (RPTPβ/ζ) family. The analyses were carried out in the retina and optic nerve of glaucomatous PTP-Meg2 HET and WT mice using quantitative real-time PCR (RT-qPCR), immunohistochemistry, and Western blot. Interestingly, we observed increased fibronectin and laminin levels in the glaucomatous HET retina and optic nerve compared to the WT group. RT-qPCR analyses of the laminins α4, β2 and γ3 showed an altered isoform-specific regulation in the HET retina and optic nerve. In addition, an upregulation of tenascin-C and its interaction partner RPTPβ/ζ/phosphacan was found in glaucomatous tissue. However, comparable protein and mRNA levels for tenascin-R as well as aggrecan and brevican were observed in both groups. Overall, our study showed a remodeling of various ECM components in the glaucomatous retina and optic nerve of PTP-Meg2 HET mice. This dysregulation could be responsible for pathological processes such as neovascularization, inflammation, and reactive gliosis in glaucomatous neurodegeneration.
Collapse
|
13
|
Rathjen FG, Hodge R. Early Days of Tenascin-R Research: Two Approaches Discovered and Shed Light on Tenascin-R. Front Immunol 2021; 11:612482. [PMID: 33488619 PMCID: PMC7820773 DOI: 10.3389/fimmu.2020.612482] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Fritz G Rathjen
- Department of Neuroscience, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Russell Hodge
- Department of Neuroscience, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
14
|
Extracellular Matrix in Neural Plasticity and Regeneration. Cell Mol Neurobiol 2020; 42:647-664. [PMID: 33128689 DOI: 10.1007/s10571-020-00986-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix (ECM) is a fundamental component of biological tissues. The ECM in the central nervous system (CNS) is unique in both composition and function. Functions such as learning, memory, synaptogenesis, and plasticity are regulated by numerous ECM molecules. The neural ECM acts as a non-specific physical barrier that modulates neuronal plasticity and axon regeneration. There are two specialized types of ECM in the CNS, diffuse perisynaptic ECM and condensed ECM, which selectively surround the perikaryon and initial part of dendritic trees in subtypes of neurons, forming perineuronal nets. This review presents the current knowledge about the role of important neuronal ECM molecules in maintaining the basic functions of a neuron, including electrogenesis and the ability to form neural circuits. The review mainly focuses on the role of ECM components that participate in the control of key events such as cell survival, axonal growth, and synaptic remodeling. Particular attention is drawn to the numerous molecular partners of the main ECM components. These regulatory molecules are integrated into the cell membrane or disposed into the matrix itself in solid or soluble form. The interaction of the main matrix components with molecular partners seems essential in molecular mechanisms controlling neuronal functions. Special attention is paid to the chondroitin sulfate proteoglycan 4, type 1 transmembrane protein, neural-glial antigen 2 (NG2/CSPG4), whose cleaved extracellular domain is such a molecular partner that it not only acts directly on neural and vascular cells, but also exerts its influence indirectly by binding to resident ECM molecules.
Collapse
|
15
|
Minta K, Cullen NC, Nimer FA, Thelin EP, Piehl F, Clarin M, Tullberg M, Jeppsson A, Portelius E, Zetterberg H, Blennow K, Andreasson U. Dynamics of extracellular matrix proteins in cerebrospinal fluid and serum and their relation to clinical outcome in human traumatic brain injury. Clin Chem Lab Med 2020; 57:1565-1573. [PMID: 30980710 DOI: 10.1515/cclm-2019-0034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/24/2019] [Indexed: 12/20/2022]
Abstract
Background Brevican, neurocan, tenascin-C and tenascin-R are extracellular matrix proteins present in brain that show increased expression in experimental animal models of brain injury. However, little is known about the dynamics of these proteins in human body fluids, such as cerebrospinal fluid (CSF) and serum, after traumatic brain injury (TBI). The aims of this study were to investigate if matrix proteins in CSF and serum are associated with functional outcome following traumatic brain injury, if their concentrations change over time and to compare their levels between brain injured patients to controls. Methods In total, 42 traumatic brain injury patients, nine healthy controls and a contrast group consisting of 38 idiopathic normal pressure hydrocephalus patients were included. Enzyme-linked immunosorbent assays (ELISAs) were used to measure the concentrations of proteins. Results Increased concentrations of brevican, tenascin-C and tenascin-R in CSF correlated with unfavourable outcome, with stronger outcome prediction ability compared to other biomarkers of brain tissue injury. CSF brevican, tenascin-R and serum neurocan gradually decreased with time (p = 0.04, p = 0.008, p = 0.005, respectively), while serum tenascin-C (p = 0.01) increased. CSF concentrations of brevican, neurocan and tenascin-R (only in time point 3) after TBI were lower than in the idiopathic normal pressure hydrocephalus group (p < 0.0001, p < 0.0001, and p = 0.0008, respectively). In serum, tenascin-C concentration was higher and neurocan lower compared to healthy controls (p = 0.02 and p = 0.0009). Conclusions These findings indicate that levels of extracellular matrix proteins are associated with clinical outcome following TBI and may act as markers for different pathophysiology than currently used protein biomarkers.
Collapse
Affiliation(s)
- Karolina Minta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Nicholas C Cullen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Faiez Al Nimer
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Eric P Thelin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, UK
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Clarin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Mats Tullberg
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anna Jeppsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
16
|
Ceci M, Mariano V, Romano N. Zebrafish as a translational regeneration model to study the activation of neural stem cells and role of their environment. Rev Neurosci 2019; 30:45-66. [PMID: 30067512 DOI: 10.1515/revneuro-2018-0020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
The review is an overview of the current knowledge of neuronal regeneration properties in mammals and fish. The ability to regenerate the damaged parts of the nervous tissue has been demonstrated in all vertebrates. Notably, fish and amphibians have the highest capacity for neurogenesis, whereas reptiles and birds are able to only regenerate specific regions of the brain, while mammals have reduced capacity for neurogenesis. Zebrafish (Danio rerio) is a promising model of study because lesions in the brain or complete cross-section of the spinal cord are followed by an effective neuro-regeneration that successfully restores the motor function. In the brain and the spinal cord of zebrafish, stem cell activity is always able to re-activate the molecular programs required for central nervous system regeneration. In mammals, traumatic brain injuries are followed by reduced neurogenesis and poor axonal regeneration, often insufficient to functionally restore the nervous tissue, while spinal injuries are not repaired at all. The environment that surrounds the stem cell niche constituted by connective tissue and stimulating factors, including pro-inflammation molecules, seems to be a determinant in triggering stem cell proliferation and/or the trans-differentiation of connective elements (mainly fibroblasts). Investigating and comparing the neuronal regeneration in zebrafish and mammals may lead to a better understanding of the mechanisms behind neurogenesis, and the failure of the regenerative response in mammals, first of all, the role of inflammation, considered the main inhibitor of the neuronal regeneration.
Collapse
Affiliation(s)
- Marcello Ceci
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| | - Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicla Romano
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| |
Collapse
|
17
|
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]
|
18
|
Chen X, Yuan R, Gao L, Huang C, Fan W, Ye J, Chen C. Induction of CRMP-2 phosphorylation by CDK5 restricts the repair of damaged optic nerve. J Cell Physiol 2018; 234:11240-11246. [PMID: 30537069 DOI: 10.1002/jcp.27778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/30/2018] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To study the mechanism of collapsin response mediator protein-2 (CRMP-2) phosphorylation changes and cyclin-dependent kinase 5 (CDK5) expression after optic nerve injury. METHODS Optic nerve injury rat models were constructed, the messenger RNA (mRNA) level of CRMP-2 in optic nerve tissues was determined by quantitative reverse transcription polymerase chain reaction (qRT-PCR) after building models 0, 3, 7, and 14 days. The protein expression of CRMP-2, phospho-CRMP-2 (p-CRMP-2), and CDK5 were also determined by western blot analysis. Lentivirus overexpressing CRMP-2 and CRMP-2 small interfering RNA (siRNA) plasmid were designed and transfected to retina ganglion cells (RGCs), and then the neurites outgrowth of RGCs were cultured with CDK5 inhibitor or CDK5 activator was determined by tubulin staining. Inhibition on CDK5 promotes injured optic nerve by using carrying CDK5 siRNA inject into vitreous chamber. RESULTS There was no significant change in CRMP-2 expression in optic nerve injury rat, while p-CRMP-2 expression was evidently increased compared with sham operation group. The expression level of CDK5 in optic nerve tissue was upregulated after optic nerve injury in rat, and the upward trend of p-CRMP-2 and CDK5 was consistent with the time after the injury was prolonged. Inhibition on CDK5 evidently decreased the expression of p-CRMP-2. CDK5 siRNA had an obvious repair effect on the injured optic nerve. CONCLUSION The increase of CDK5 activity can lead to CRMP-2 hyperphosphorylation, which results in the difficult repair of damaged optic nerve. Therefore, inhibition on CDK5 could promote the repair of damaged optic nerve.
Collapse
Affiliation(s)
- Xiaofan Chen
- Department of Ophthalmology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Rongdi Yuan
- Department of Ophthalmology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Ling Gao
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Chanjuan Huang
- Department of Ophthalmology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Wei Fan
- Department of Ophthalmology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Chunlin Chen
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
19
|
Li HJ, Sun ZL, Yang XT, Zhu L, Feng DF. Exploring Optic Nerve Axon Regeneration. Curr Neuropharmacol 2018; 15:861-873. [PMID: 28029073 PMCID: PMC5652030 DOI: 10.2174/1570159x14666161227150250] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 12/14/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022] Open
Abstract
Background: Traumatic optic nerve injury is a leading cause of irreversible blindness across the world and causes progressive visual impairment attributed to the dysfunction and death of retinal ganglion cells (RGCs). To date, neither pharmacological nor surgical interventions are sufficient to halt or reverse the progress of visual loss. Axon regeneration is critical for functional recovery of vision following optic nerve injury. After optic nerve injury, RGC axons usually fail to regrow and die, leading to the death of the RGCs and subsequently inducing the functional loss of vision. However, the detailed molecular mechanisms underlying axon regeneration after optic nerve injury remain poorly understood. Methods: Research content related to the detailed molecular mechanisms underlying axon regeneration after optic nerve injury have been reviewed. Results: The present review provides an overview of regarding potential strategies for axonal regeneration of RGCs and optic nerve repair, focusing on the role of cytokines and their downstream signaling pathways involved in intrinsic growth program and the inhibitory environment together with axon guidance cues for correct axon guidance. A more complete understanding of the factors limiting axonal regeneration will provide a rational basis, which contributes to develop improved treatments for optic nerve regeneration. These findings are encouraging and open the possibility that clinically meaningful regeneration may become achievable in the future. Conclusion: Combination of treatments towards overcoming growth-inhibitory molecules and enhancing intrinsic growth capacity combined with correct guidance using axon guidance cues is crucial for developing promising therapies to promote axon regeneration and functional recovery after ON injury.
Collapse
Affiliation(s)
- Hong-Jiang Li
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Zhao-Liang Sun
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Xi-Tao Yang
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Liang Zhu
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Dong-Fu Feng
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| |
Collapse
|
20
|
Zuliani-Alvarez L, Marzeda AM, Deligne C, Schwenzer A, McCann FE, Marsden BD, Piccinini AM, Midwood KS. Mapping tenascin-C interaction with toll-like receptor 4 reveals a new subset of endogenous inflammatory triggers. Nat Commun 2017; 8:1595. [PMID: 29150600 PMCID: PMC5693923 DOI: 10.1038/s41467-017-01718-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/05/2017] [Indexed: 02/08/2023] Open
Abstract
Pattern recognition underpins innate immunity; the accurate identification of danger, including infection, injury, or tumor, is key to an appropriately targeted immune response. Pathogen detection is increasingly well defined mechanistically, but the discrimination of endogenous inflammatory triggers remains unclear. Tenascin-C, a matrix protein induced upon tissue damage and expressed by tumors, activates toll-like receptor 4 (TLR4)-mediated sterile inflammation. Here we map three sites within tenascin-C that directly and cooperatively interact with TLR4. We also identify a conserved inflammatory epitope in related proteins from diverse families, and demonstrate that its presence targets molecules for TLR detection, while its absence enables escape of innate immune surveillance. These data reveal a unique molecular code that defines endogenous proteins as inflammatory stimuli by marking them for recognition by TLRs.
Collapse
Affiliation(s)
- Lorena Zuliani-Alvarez
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Anna M Marzeda
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Claire Deligne
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Anja Schwenzer
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Fiona E McCann
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Brian D Marsden
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK.,Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7DQ, UK
| | - Anna M Piccinini
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK.,School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Kim S Midwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK.
| |
Collapse
|
21
|
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
|
22
|
Ischemic injury leads to extracellular matrix alterations in retina and optic nerve. Sci Rep 2017; 7:43470. [PMID: 28262779 PMCID: PMC5338032 DOI: 10.1038/srep43470] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/25/2017] [Indexed: 01/10/2023] Open
Abstract
Retinal ischemia occurs in a variety of eye diseases. Restrained blood flow induces retinal damage, which leads to progressive optic nerve degeneration and vision loss. Previous studies indicate that extracellular matrix (ECM) constituents play an important role in complex tissues, such as retina and optic nerve. They have great impact on de- and regeneration processes and represent major candidates of central nervous system glial scar formation. Nevertheless, the importance of the ECM during ischemic retina and optic nerve neurodegeneration is not fully understood yet. In this study, we analyzed remodeling of the extracellular glycoproteins fibronectin, laminin, tenascin-C and tenascin-R and the chondroitin sulfate proteoglycans (CSPGs) aggrecan, brevican and phosphacan/RPTPβ/ζ in retinae and optic nerves of an ischemia/reperfusion rat model via quantitative real-time PCR, immunohistochemistry and Western blot. A variety of ECM constituents were dysregulated in the retina and optic nerve after ischemia. Regarding fibronectin, significantly elevated mRNA and protein levels were observed in the retina following ischemia, while laminin and tenascin-C showed enhanced immunoreactivity in the optic nerve after ischemia. Interestingly, CSPGs displayed significantly increased expression levels in the optic nerve. Our study demonstrates a dynamic expression of ECM molecules following retinal ischemia, which strengthens their regulatory role during neurodegeneration.
Collapse
|
23
|
Hyper-IL-6: a potent and efficacious stimulator of RGC regeneration. Eye (Lond) 2016; 31:173-178. [PMID: 27886185 DOI: 10.1038/eye.2016.234] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 09/29/2016] [Indexed: 01/11/2023] Open
Abstract
Mature retinal ganglion cells (RGCs) normally fail to regenerate injured axons and die soon after optic nerve injury. Research over the last two decades has demonstrated that application of IL-6-like cytokines or activation of respective downstream signaling pathways promote neuroprotection and optic nerve regeneration. However, the overall beneficial effects of natural cytokines remain usually rather moderate, possibly due to intrinsic signaling pathway inhibitors, such as PTEN or SOCS3, or a limited expression of specific cytokine receptors in RGCs. It was recently demonstrated that directly targeting the gp130 receptor, a common signalling receptor of all IL-6-like cytokines, induces stronger RGC axon regeneration in vitro and in vivo than other known growth-promoting treatments such as inflammatory stimulation or PTEN knockout. Remarkably, continuous expression of hyper-IL-6 (hIL-6) upon intravitreal AAV injection after nerve injury enables long-distance axon regeneration, with some axons growing through the optic chiasm 6 weeks after optic nerve injury. Thus, AAV-mediated hIL-6 delivery is so far one of the strongest single, post-injury treatments for the promotion of optic nerve regeneration and may be suitable for the development of novel, clinically applicable therapeutic treatments for human patients.
Collapse
|
24
|
Ahuja CS, Fehlings M. Concise Review: Bridging the Gap: Novel Neuroregenerative and Neuroprotective Strategies in Spinal Cord Injury. Stem Cells Transl Med 2016; 5:914-24. [PMID: 27130222 DOI: 10.5966/sctm.2015-0381] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/07/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Spinal cord injuries (SCIs) result in devastating lifelong disability for patients and their families. The initial mechanical trauma is followed by a damaging secondary injury cascade involving proapoptotic signaling, ischemia, and inflammatory cell infiltration. Ongoing cellular necrosis releases ATP, DNA, glutamate, and free radicals to create a cytotoxic postinjury milieu. Long-term regeneration of lost or injured networks is further impeded by cystic cavitation and the formation of an inhibitory glial-chondroitin sulfate proteoglycan scar. In this article, we discuss important neuroprotective interventions currently applied in clinical practice, including surgical decompression, blood pressure augmentation, and i.v. methylprednisolone. We then explore exciting translational therapies on the horizon, such as riluzole, minocycline, fibroblast growth factor, magnesium, and hypothermia. Finally, we summarize the key neuroregenerative strategies of the next decade, including glial scar degradation, Rho-ROCK inhibition, cell-based therapies, and novel bioengineered adjuncts. Throughout, we emphasize the need for combinatorial approaches to this multifactorial problem and discuss relevant studies at the forefront of translation. We conclude by providing our perspectives on the future direction of SCI research. SIGNIFICANCE Spinal cord injuries (SCIs) result in devastating, lifelong disability for patients and their families. This article discusses important neuroprotective interventions currently applied in clinical practice, including surgical decompression, blood pressure augmentation, and i.v. methylprednisolone. Translational therapies on the horizon are discussed, such as riluzole, minocycline, fibroblast growth factor, magnesium, and hypothermia. The key neuroregenerative strategies of the next decade are summarized, including glial scar degradation, Rho-ROCK inhibition, cell-based therapies, and novel bioengineered adjuncts. The need for combinatorial approaches to this multifactorial problem is emphasized, relevant studies at the forefront of translation are discussed, and perspectives on the future direction of SCI research are presented.
Collapse
Affiliation(s)
- Christopher S Ahuja
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Michael Fehlings
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada McEwen Centre for Regenerative Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada Department of Surgery, University of Toronto, Toronto, Ontario, Canada Spine Program, University of Toronto, Toronto, Ontario, Canada McLaughlin Centre for Molecular Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Dawson AJ, Miotke JA, Meyer RL. Intraocular BDNF promotes ectopic branching, alters motility and stimulates abnormal collaterals in regenerating optic fibers. Brain Res 2015; 1613:13-26. [PMID: 25847715 DOI: 10.1016/j.brainres.2015.03.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 03/24/2015] [Accepted: 03/26/2015] [Indexed: 01/02/2023]
Abstract
A great deal of effort has been invested in using trophic factors and other bioactive molecules to promote cell survival and axonal regeneration in the adult central nervous system. Far less attention has been paid to investigating potential effects that trophic factors may have that might interfere with recovery. In the visual system, BDNF has been previously reported to prevent regeneration. To test if BDNF is inherently incompatible with regeneration, BDNF was given intraocularly during optic nerve regeneration in the adult goldfish. In vivo imaging and anatomical analysis of selectively labeled axons were used as a sensitive assay for effects on regeneration within the tectum. BDNF had no detectable inhibitory effect on the ability of axons to regenerate. Normal numbers of axons regenerated into the tectum, exhibited dynamic growth and retractions similar to controls, and were able to navigate to their correct target zone in the tectum. However, BDNF was found to have additional effects that adversely affected the quality of regeneration. It promoted premature branching at ectopic locations, diminished the growth rate of axons through the tectum, and resulted in the formation of ectopic collaterals. Thus, although BDNF has robust effects on axonal behavior, it is, nevertheless, compatible with axonal regeneration, axon navigation and the formation of terminal arbors.
Collapse
Affiliation(s)
- Amy J Dawson
- Department of Developmental and Cell Biology, University of California at Irvine, Irvine, CA 92697, USA; Division of Arts and Sciences, New River Community College, Dublin, VA 24084, USA.
| | - Jill A Miotke
- Department of Developmental and Cell Biology, University of California at Irvine, Irvine, CA 92697, USA.
| | - Ronald L Meyer
- Department of Developmental and Cell Biology, University of California at Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
26
|
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]
|
27
|
|
28
|
Gaudet AD, Popovich PG. Extracellular matrix regulation of inflammation in the healthy and injured spinal cord. Exp Neurol 2014; 258:24-34. [PMID: 25017885 DOI: 10.1016/j.expneurol.2013.11.020] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 02/06/2023]
Abstract
Throughout the body, the extracellular matrix (ECM) provides structure and organization to tissues and also helps regulate cell migration and intercellular communication. In the injured spinal cord (or brain), changes in the composition and structure of the ECM undoubtedly contribute to regeneration failure. Less appreciated is how the native and injured ECM influences intraspinal inflammation and, conversely, how neuroinflammation affects the synthesis and deposition of ECM after CNS injury. In all tissues, inflammation can be initiated and propagated by ECM disruption. Molecules of ECM newly liberated by injury or inflammation include hyaluronan fragments, tenascins, and sulfated proteoglycans. These act as "damage-associated molecular patterns" or "alarmins", i.e., endogenous proteins that trigger and subsequently amplify inflammation. Activated inflammatory cells, in turn, further damage the ECM by releasing degradative enzymes including matrix metalloproteinases (MMPs). After spinal cord injury (SCI), destabilization or alteration of the structural and chemical compositions of the ECM affects migration, communication, and survival of all cells - neural and non-neural - that are critical for spinal cord repair. By stabilizing ECM structure or modifying their ability to trigger the degradative effects of inflammation, it may be possible to create an environment that is more conducive to tissue repair and axon plasticity after SCI.
Collapse
Affiliation(s)
- Andrew D Gaudet
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, College of Medicine, The Ohio State University, 670 Biomedical Research Tower, 460 West 12th Ave., Columbus, OH 43210, USA.
| | - Phillip G Popovich
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, College of Medicine, The Ohio State University, 670 Biomedical Research Tower, 460 West 12th Ave., Columbus, OH 43210, USA.
| |
Collapse
|
29
|
Roll L, Faissner A. Influence of the extracellular matrix on endogenous and transplanted stem cells after brain damage. Front Cell Neurosci 2014; 8:219. [PMID: 25191223 PMCID: PMC4137450 DOI: 10.3389/fncel.2014.00219] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/18/2014] [Indexed: 01/07/2023] Open
Abstract
The limited regeneration capacity of the adult central nervous system (CNS) requires strategies to improve recovery of patients. In this context, the interaction of endogenous as well as transplanted stem cells with their environment is crucial. An understanding of the molecular mechanisms could help to improve regeneration by targeted manipulation. In the course of reactive gliosis, astrocytes upregulate Glial fibrillary acidic protein (GFAP) and start, in many cases, to proliferate. Beside GFAP, subpopulations of these astroglial cells coexpress neural progenitor markers like Nestin. Although cells express these markers, the proportion of cells that eventually give rise to neurons is limited in many cases in vivo compared to the situation in vitro. In the first section, we present the characteristics of endogenous progenitor-like cells and discuss the differences in their neurogenic potential in vitro and in vivo. As the environment plays an important role for survival, proliferation, migration, and other processes, the second section of the review describes changes in the extracellular matrix (ECM), a complex network that contains numerous signaling molecules. It appears that signals in the damaged CNS lead to an activation and de-differentiation of astrocytes, but do not effectively promote neuronal differentiation of these cells. Factors that influence stem cells during development are upregulated in the damaged brain as part of an environment resembling a stem cell niche. We give a general description of the ECM composition, with focus on stem cell-associated factors like the glycoprotein Tenascin-C (TN-C). Stem cell transplantation is considered as potential treatment strategy. Interaction of transplanted stem cells with the host environment is critical for the outcome of stem cell-based therapies. Possible mechanisms involving the ECM by which transplanted stem cells might improve recovery are discussed in the last section.
Collapse
Affiliation(s)
- Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany ; International Graduate School of Neuroscience, Ruhr-University Bochum Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany ; International Graduate School of Neuroscience, Ruhr-University Bochum Bochum, Germany
| |
Collapse
|
30
|
Distribution of extracellular matrix macromolecules in the vestibular nuclei and cerebellum of the frog, Rana esculenta. Neuroscience 2014; 258:162-73. [DOI: 10.1016/j.neuroscience.2013.10.080] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 10/31/2013] [Accepted: 10/31/2013] [Indexed: 12/31/2022]
|
31
|
|
32
|
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]
|
33
|
You J, Hong SQ, Zhang MY, Zhao HL, Liu TZ, Zhou HL, Cai YQ, Xu ZM, Guo Y, Jiang XD, Xu RX. Passive immunization with tenascin-R (TN-R) polyclonal antibody promotes axonal regeneration and functional recovery after spinal cord injury in rats. Neurosci Lett 2012; 525:129-34. [PMID: 22902990 DOI: 10.1016/j.neulet.2012.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 07/15/2012] [Accepted: 08/02/2012] [Indexed: 11/24/2022]
Abstract
Tenascin-R (TN-R) is a neural specific protein and an important molecule involved in inhibition of axonal regeneration after spinal cord injury (SCI). Here we report on rabbit-derived TN-R polyclonal antibody, which acts as a TN-R antagonist with high titer and high specificity, promoted neurite outgrowth and sprouting of rat cortical neurons cultured on the inhibitory TN-R substrate in vitro. When locally administered into the lesion sites of rats received spinal cord dorsal hemisection, these TN-R antibodies could significantly decrease RhoA activation and improve functional recovery from corticospinal tract (CST) transection. Thus, passive immunotherapy with specific TN-R antagonist may represent a promising repair strategy following acute SCI.
Collapse
Affiliation(s)
- Jian You
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Fischer D, Leibinger M. Promoting optic nerve regeneration. Prog Retin Eye Res 2012; 31:688-701. [PMID: 22781340 DOI: 10.1016/j.preteyeres.2012.06.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 06/13/2012] [Accepted: 06/21/2012] [Indexed: 10/28/2022]
Abstract
Vision is the most important sense for humans and it is irreversibly impaired by axonal damage of retinal ganglion cells (RGCs) in the optic nerve due to the lack of axonal regeneration. The failure of regeneration is partially attributable to factors located in the inhibitory environment of the forming glial scar and myelin as well as an insufficient intrinsic ability for axonal regrowth. Moreover, RGCs undergo apoptotic cell death after optic nerve injury, eliminating any chance for regeneration. In this review, we discuss the different aspects that cause regenerative failure in the optic nerve. Moreover, we describe discoveries of the last two decades demonstrating that under certain circumstances mature RGCs can be transformed into an active regenerative state allowing these neurons to survive axotomy and to regenerate axons in the injured optic nerve. In this context we focus on the role of the cytokines ciliary neutrophic factor (CNTF) and leukemia inhibitory factor (LIF), their receptors and the downstream signaling pathways. Furthermore, we discuss strategies to overcome inhibitory signaling induced by molecules associated with optic nerve myelin and the glial scar as well as the regenerative outcome after combinatorial treatments. These findings are encouraging and may open the possibility that clinically meaningful regeneration may become achievable one day in the future.
Collapse
Affiliation(s)
- Dietmar Fischer
- Department of Neurology, Experimental Neurology, Heinrich Heine University Düsseldorf, Merowingerplatz 1a, 40225 Düsseldorf, Germany.
| | | |
Collapse
|
35
|
Franke H, Verkhratsky A, Burnstock G, Illes P. Pathophysiology of astroglial purinergic signalling. Purinergic Signal 2012; 8:629-57. [PMID: 22544529 DOI: 10.1007/s11302-012-9300-0] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/01/2012] [Indexed: 12/13/2022] Open
Abstract
Astrocytes are fundamental for central nervous system (CNS) physiology and are the fulcrum of neurological diseases. Astroglial cells control development of the nervous system, regulate synaptogenesis, maturation, maintenance and plasticity of synapses and are central for nervous system homeostasis. Astroglial reactions determine progression and outcome of many neuropathologies and are critical for regeneration and remodelling of neural circuits following trauma, stroke, ischaemia or neurodegenerative disorders. They secrete multiple neurotransmitters and neurohormones to communicate with neurones, microglia and the vascular walls of capillaries. Signalling through release of ATP is the most widespread mean of communication between astrocytes and other types of neural cells. ATP serves as a fast excitatory neurotransmitter and has pronounced long-term (trophic) roles in cell proliferation, growth, and development. During pathology, ATP is released from damaged cells and acts both as a cytotoxic factor and a proinflammatory mediator, being a universal "danger" signal. In this review, we summarise contemporary knowledge on the role of purinergic receptors (P2Rs) in a variety of diseases in relation to changes of astrocytic functions and nucleotide signalling. We have focussed on the role of the ionotropic P2X and metabotropic P2YRs working alone or in concert to modify the release of neurotransmitters, to activate signalling cascades and to change the expression levels of ion channels and protein kinases. All these effects are of great importance for the initiation, progression and maintenance of astrogliosis-the conserved and ubiquitous glial defensive reaction to CNS pathologies. We highlighted specific aspects of reactive astrogliosis, especially with respect to the involvement of the P2X(7) and P2Y(1)R subtypes. Reactive astrogliosis exerts both beneficial and detrimental effects in a context-specific manner determined by distinct molecular signalling cascades. Understanding the role of purinergic signalling in astrocytes is critical to identifying new therapeutic principles to treat acute and chronic neurological diseases.
Collapse
Affiliation(s)
- Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstrasse 16-18, 04107, Leipzig, Germany.
| | | | | | | |
Collapse
|
36
|
Blockade of P2 nucleotide receptors after spinal cord injury reduced the gliotic response and spared tissue. J Mol Neurosci 2011; 46:167-76. [PMID: 21647706 DOI: 10.1007/s12031-011-9567-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 05/23/2011] [Indexed: 12/22/2022]
Abstract
Spinal cord injury (SCI) triggers a sequel of events commonly associated with cell death and dysfunction of glias and neurons surrounding the lesion. Although astrogliosis and glial scar formation have been involved in both damage and repair processes after SCI, their role remains controversial. Our goal was to investigate the effects of the P2 receptors antagonists, PPADS and suramin, in the establishment of the reactive gliosis and the formation of the glial scar. Molecular biology, immunohistochemistry, spared tissue, and locomotor behavioral studies were used to evaluate astrogliosis, in adult female Sprague-Dawley rats treated with P2 antagonists after moderate injury with the NYU impactor device. Semi-quantitative RT-PCR confirmed the presence of P2Y(1,) P2Y(2,) P2Y(4,) P2Y(6,) P2Y(12), and P2X(2) receptors in the adult spinal cord. Immunohistochemistry studies confirmed a significant decrease in GFAP-labeled cells at the injury epicenter as well as a decrease in spared tissue after treatment with the antagonists. Functional open field testing revealed no significant locomotor score differences between treated and control animals. Our work is consistent with studies suggesting that astrogliosis is an important event after SCI that limits tissue damage and lesion spreading.
Collapse
|
37
|
Fleisch VC, Fraser B, Allison WT. Investigating regeneration and functional integration of CNS neurons: lessons from zebrafish genetics and other fish species. Biochim Biophys Acta Mol Basis Dis 2010; 1812:364-80. [PMID: 21044883 DOI: 10.1016/j.bbadis.2010.10.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 10/05/2010] [Accepted: 10/21/2010] [Indexed: 12/21/2022]
Abstract
Zebrafish possess a robust, innate CNS regenerative ability. Combined with their genetic tractability and vertebrate CNS architecture, this ability makes zebrafish an attractive model to gain requisite knowledge for clinical CNS regeneration. In treatment of neurological disorders, one can envisage replacing lost neurons through stem cell therapy or through activation of latent stem cells in the CNS. Here we review the evidence that radial glia are a major source of CNS stem cells in zebrafish and thus activation of radial glia is an attractive therapeutic target. We discuss the regenerative potential and the molecular mechanisms thereof, in the zebrafish spinal cord, retina, optic nerve and higher brain centres. We evaluate various cell ablation paradigms developed to induce regeneration, with particular emphasis on the need for (high throughput) indicators that neuronal regeneration has restored sensory or motor function. We also examine the potential confound that regeneration imposes as the community develops zebrafish models of neurodegeneration. We conclude that zebrafish combine several characters that make them a potent resource for testing hypotheses and discovering therapeutic targets in functional CNS regeneration. This article is part of a Special Issue entitled Zebrafish Models of Neurological Diseases.
Collapse
Affiliation(s)
- Valerie C Fleisch
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, Alberta, Canada.
| | | | | |
Collapse
|
38
|
Rodríguez-Zayas AE, Torrado AI, Miranda JD. P2Y2 receptor expression is altered in rats after spinal cord injury. Int J Dev Neurosci 2010; 28:413-21. [PMID: 20619335 DOI: 10.1016/j.ijdevneu.2010.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 06/22/2010] [Accepted: 07/01/2010] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury increases inhibitory factors that may restrict neurite outgrowth after trauma. The expression of repulsive molecules in reactive astrocytes and the formation of the glial scar at the injury site produce the non-permissive environment for axonal regeneration. However, the mechanism that triggers this astrogliotic response is unknown. The release of nucleotides has been linked to this hypertrophic state. Our goal is to investigate the temporal profile of P2Y(2) nucleotide receptor after spinal cord injury in adult female Sprague-Dawley rats. Molecular biology, immunofluorescence studies, and Western Blots were used to evaluate the temporal profile (2, 4, 7, 14, and 28 days post-injury) of this receptor in rats injured at the T-10 level using the NYU impactor device. Real time RT-PCR showed a significant increase of P2Y(2) mRNA after 2 days post-injury that continues throughout 28 days post-injury. Double labeling studies localized P2Y(2) immunoreactivity in neuronal cell bodies, axons, macrophages, oligodendrocytes and reactive astrocytes. Immunofluorescence studies also demonstrated a low level of P2Y(2) receptor in sham samples, which increased after injury in glial fibrillary acidic protein positive cells. Western Blot performed with contused spinal cord protein samples revealed an upregulation in the P2Y(2) 42 kDa protein band expression after 4 days post-injury that continues until 28 days post-injury. However, a downregulation of the 62 kDa receptor protein band after 2 days post-injury that continues up to 28 days post-injury was observed. Therefore, the spatio-temporal pattern of P2Y(2) gene expression after spinal cord injury suggests a role in the pathophysiology response generated after trauma.
Collapse
Affiliation(s)
- Ana E Rodríguez-Zayas
- Department of Physiology, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico
| | | | | |
Collapse
|
39
|
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
|
40
|
Manor T, Barbiro-Michaely E, Rogatsky G, Mayevsky A. Real-time multi-site multi-parametric monitoring of rat brain subjected to traumatic brain injury. Neurol Res 2008; 30:1075-83. [PMID: 18826757 DOI: 10.1179/174313208x346107] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is one of the major causes of death in the world, with at least ten million serious traumatic brain injuries occurring annually; nevertheless, the pathophysiologic events taking place immediately after the injury are not yet fully known. OBJECTIVE To study the effects of TBI on brain hemodynamic, metabolic and ionic homeostasis using the multi-parametric monitoring system. This system enables real-time monitoring of cerebral blood flow (CBF), mitochondrial NADH redox state, extracellular levels of K+, H+, DC potential, ECoG and ICP. METHODS In order to find the best brain location for the monitoring device in relation to the fluid percussion injury site, we used the multi-site multi-parametric monitoring system. Two groups of rats were connected to four monitoring probes at four different locations near the injury site, two in each hemisphere. We monitored CBF, NADH redox state, tissue reflectance and DC steady potential in each of the four sites. RESULTS Under anoxia, the initial CBF decrease was followed by an increase, NADH level increased, the reflectance decreased and dc potential showed a biphasic response, in all 4 locations. However, following fluid percussion injury, there was a significant variability in the responses in each of the 4 monitored locations. CONCLUSION The advantage of the multi-parametric-monitoring approach for enhanced understanding of the injured brain was indicated. Moreover, we showed that contralateral monitoring of the injured brain gives good indication for the events taking place following fluid percussion brain injury.
Collapse
Affiliation(s)
- Tami Manor
- Mina and Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 52900, Israel
| | | | | | | |
Collapse
|
41
|
Becker CG, Becker T. Growth and pathfinding of regenerating axons in the optic projection of adult fish. J Neurosci Res 2008; 85:2793-9. [PMID: 17131420 DOI: 10.1002/jnr.21121] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In contrast to mammals, teleost fish are able to regrow severed long-range projection axons in the central nervous system (CNS), leading to recovery of function. The optic projection in teleost fish is used to study neuron-intrinsic and environmental molecular factors that determine successful axon regrowth and navigation through a complex CNS pathway back to original targets. Here we review evidence for regeneration-specific regulation and robust expression of growth- and pathfinding-associated genes in regenerating retinal ganglion cell (RGC) axons of adult fish. The environment of the CNS in fish appears to contain few inhibitory molecules and at the same time a number of promoting molecules for axon regrowth. Finally, some environmental cues that are used as guidance cues for developing RGC axons are also present in continuously growing adult animals. These molecules may serve as guidance cues for the precise navigation of axons from newly generated RGCs in adult animals as well as of regenerating RGC axons after a lesion. The application of new molecular techniques especially to adult zebrafish, is likely to produce new insights into successful axonal regeneration in the CNS of fish and the absence thereof in mammals.
Collapse
Affiliation(s)
- Catherina G Becker
- Centre for Neuroscience Research, University of Edinburgh, Summerhall, Edinburgh, United Kingdom
| | | |
Collapse
|
42
|
Lang DM, Monzon-Mayor M, del Mar Romero-Aleman M, Yanes C, Santos E, Pesheva P. Tenascin-R and axon growth-promoting molecules are up-regulated in the regenerating visual pathway of the lizard (Gallotia galloti). Dev Neurobiol 2008; 68:899-916. [DOI: 10.1002/dneu.20624] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
43
|
Cheema SK, Chen E, Shea LD, Mathur AB. Regulation and guidance of cell behavior for tissue regeneration via the siRNA mechanism. Wound Repair Regen 2007; 15:286-95. [PMID: 17537114 DOI: 10.1111/j.1524-475x.2007.00228.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
RNA intereference and short-interfering RNA (siRNA) have been proven to be effective at decreasing the expression of target genes and provide a valuable tool for promoting and directing the growth of functional tissues for repair and reconstructive tissue engineering applications. siRNA is a gene-silencing mechanism that involves double-stranded RNA-mediated sequence-specific mRNA degradation and is a powerful mechanism for controlling cell behavior. The use of siRNA to reduce the expression of a target gene can induce the expression of one or more tissue-inductive factors, direct the differentiation of stem or progenitor cells, or remove a factor that inhibits regeneration, which can be useful in fundamental studies of tissue formation or in applications to promote in vivo regeneration. The potential of siRNA is illustrated through specific examples within the fields of angiogenesis, bone and nerve regeneration, and wound healing. In addition, challenges to deliver siRNA effectively for tissue engineering applications are addressed. siRNA represents a powerful tool to investigate and/or promote tissue formation, and numerous opportunities exist for identifying targets that promote regeneration of tissue and developing effective delivery systems.
Collapse
Affiliation(s)
- Sangeeta K Cheema
- Department of Plastic Surgery, and Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77230-1402, USA
| | | | | | | |
Collapse
|
44
|
Abstract
Despite a relatively long history, general knowledge is not widespread that adult neurons can be maintained in cell culture for fairly extended periods of time. Within the central nervous system, this capacity seems to be particularly well developed in the retina, although it is still not clear whether this property is due to physical reasons (spatial configuration, simple connections) or to more fundamental differences (molecular composition, physiological function). Irrespective of the reasons, in vitro model systems are useful for investigating physiological and pathological processes occurring in mature retina. The authors argue that the numerous molecular changes undergone during maturation (modifications in ion channels and receptors, apoptotic pathways and growth factor effects) should be taken into account when using in vitro approaches to study processes involved in photoreceptor and ganglion cell degeneration, and hence that more classical methods relying on embryonic or newborn tissue should be interpreted with caution. A number of examples are given where the use of adult retinal neuronal culture may be especially informative: neurite regeneration, neuroprotection assays and pathogenic mechanisms; and areas of further research that should be explored: cell transplantation.
Collapse
Affiliation(s)
- Carl Romano
- Retina Discovery, Alcon Laboratories Inc., 6201 South Freeway, Fort Worth, TX 76134-2099, USA
| | | |
Collapse
|
45
|
Hermanns S, Klapka N, Gasis M, Müller HW. The collagenous wound healing scar in the injured central nervous system inhibits axonal regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:177-90. [PMID: 16955711 DOI: 10.1007/0-387-30128-3_11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Following traumatic injuries of the central nervous system (CNS) a wound healing scar, resembling the molecular structure of a basement membrane and mainly composed of Collagen type IV and associated glycoproteins and proteoglycans, is formed. It is well known that CNS fibers poorly regenerate after traumatic injuries. In this article we summarize data showing that prevention of collagen scar formation enables severed axons in brain and spinal cord to regrow across the lesion site and to elongate in uninjured CNS tissue. We observed that regenerating fibers grow back to their former target where they develop chemical synapses, become remyelinated by resident oligodendrocytes and conduct action potentials.
Collapse
|
46
|
Sarikcioglu L, Demir N, Demirtop A. A standardized method to create optic nerve crush: Yasargil aneurysm clip. Exp Eye Res 2006; 84:373-7. [PMID: 17157296 DOI: 10.1016/j.exer.2006.10.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Revised: 09/04/2006] [Accepted: 10/18/2006] [Indexed: 10/23/2022]
Abstract
It is often difficult to compare results obtained by different investigators on nerve compression injuries, owing to differences in method of pressure application and noncomparable pressure levels. In the present study, we described a new method to crush the optic nerve by using a specially designed and commercially available device. We think that standardization of the compression methods is necessary to compare interlaboratory results.
Collapse
Affiliation(s)
- Levent Sarikcioglu
- Department of Anatomy, Akdeniz University, Faculty of Medicine, 07070 Antalya, Turkey.
| | | | | |
Collapse
|
47
|
Apostolova I, Irintchev A, Schachner M. Tenascin-R restricts posttraumatic remodeling of motoneuron innervation and functional recovery after spinal cord injury in adult mice. J Neurosci 2006; 26:7849-59. [PMID: 16870730 PMCID: PMC6674222 DOI: 10.1523/jneurosci.1526-06.2006] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Tenascin-R (TNR) is an extracellular glycoprotein in the CNS implicated in neural development and plasticity. Its repellent properties for growing axons in a choice situation with a conducive substrate in vitro have indicated that TNR may impede regeneration in the adult mammalian CNS. Here we tested whether constitutive lack of TNR has beneficial impacts on recovery from spinal cord injury in adult mice. Using the Basso, Beattie, Bresnahan (BBB) locomotor rating scale, we found that open-field locomotion in TNR-deficient (TNR-/-) mice recovered better that in wild-type (TNR+/+) littermates after compression of the thoracic spinal cord. We also designed, validated, and applied a motion analysis approach allowing numerical assessment of motor functions. We found, in agreement with the BBB score, that functions requiring low levels of supraspinal control such as plantar stepping improved more in TNR-/- mice. This was not the case for motor tasks demanding precision such as ladder climbing. Morphological analyses revealed no evidence that improved recovery of some functions in the mutant mice were attributable to enhanced tissue sparing or axonal regrowth. Estimates of perisomatic puncta revealed reduced innervation by cholinergic and GABAergic terminals around motoneurons in intact TNR-/- compared with TNR+/+ mice. Relative to nonlesioned animals, spinal cord repair was associated with increase in GABAergic and decrease of glutamatergic puncta in TNR-/- but not in TNR+/+ mice. Our results suggest that TNR restricts functional recovery by limiting posttraumatic remodeling of synapses around motoneuronal cell bodies where TNR is normally expressed in perineuronal nets.
Collapse
|
48
|
Pesheva P, Probstmeier R, Lang DM, McBride R, Hsu NJ, Gennarini G, Spiess E, Peshev Z. Early coevolution of adhesive but not antiadhesive tenascin-R ligand-receptor pairs in vertebrates: A phylogenetic study. Mol Cell Neurosci 2006; 32:366-86. [PMID: 16831557 DOI: 10.1016/j.mcn.2006.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Revised: 05/23/2006] [Accepted: 05/25/2006] [Indexed: 01/25/2023] Open
Abstract
Axon growth inhibitory CNS matrix proteins, such as tenascin-R (TN-R), have been supposed to contribute to the poor regenerative capacity of adult mammalian CNS. With regard to TN-R function in low vertebrates capable of CNS regeneration, questions of particular interest concern the (co)evolution of ligand-receptor pairs and cellular response mechanisms associated with axon growth inhibition and oligodendrocyte differentiation. We address here these questions in a series of comparative in vivo and in vitro analyses using TN-R proteins purified from different vertebrates (from fish to human). Our studies provide strong evidence that unlike TN-R of higher vertebrates, fish TN-R proteins are not repellent for fish and less repellent for mammalian neurons and do not interfere with F3/contactin- and fibronectin-mediated mammalian cell adhesion and axon growth. However, axonal repulsion is induced in fish neurons by mammalian TN-R proteins, suggesting that the intracellular inhibitory machinery induced by TN-R-F3 interactions is already present during early vertebrate evolution. In contrast to TN-R-F3, TN-R-sulfatide interactions, mediating oligodendrocyte adhesion and differentiation, are highly conserved during vertebrate evolution. Our findings thus indicate the necessity of being cautious about extrapolations of the function of ligand-receptor pairs beyond a species border and, therefore, about the phylogenetic conservation of a molecular function at the cellular/tissue level.
Collapse
Affiliation(s)
- Penka Pesheva
- Neuro- and Tumor Cell Biology Group, Department of Nuclear Medicine, University of Bonn, Sigmund Freud Str. 25, 53105 Bonn, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Becker CG, Schweitzer J, Feldner J, Schachner M, Becker T. Tenascin-R as a repellent guidance molecule for newly growing and regenerating optic axons in adult zebrafish. Mol Cell Neurosci 2004; 26:376-89. [PMID: 15234343 DOI: 10.1016/j.mcn.2004.03.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2004] [Revised: 02/23/2004] [Accepted: 03/03/2004] [Indexed: 10/26/2022] Open
Abstract
In adult fish, in contrast to mammals, new optic axons are continuously added to the optic projection, and optic axons regrow after injury. Thus, pathfinding of optic axons during development, adult growth, and adult regeneration may rely on the same guidance cues. We have shown that tenascin-R, a component of the extracellular matrix, borders the optic pathway in developing zebrafish and acts as a repellent guidance molecule for optic axons. Here we analyze tenascin-R expression patterns along the unlesioned and lesioned optic pathway of adult zebrafish and test the influence of tenascin-R on growing optic axons of adult fish in vitro. Within intraretinal fascicles of optic axons and in the optic nerve, newly added optic axons grow in a tenascin-R immunonegative pathway, which is bordered by tenascin-R immunoreactivity. In the brain, tenascin-R expression domains in the ventral diencephalon, in non-retinorecipient pretectal nuclei and in some tectal layers closely border the optic pathway in unlesioned animals and during axon regrowth. We mimicked these boundary situations with a sharp substrate border of tenascin-R in vitro. Optic axons emanating from adult retinal explants were repelled by tenascin-R substrate borders. This is consistent with a function of tenascin-R as a repellent guidance molecule in boundaries for adult optic axons. Thus, tenascin-R may guide newly added and regenerating optic axons by a contact-repellent mechanism in the optic pathway of adult fish.
Collapse
Affiliation(s)
- Catherina G Becker
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, D-20246 Hamburg, Germany.
| | | | | | | | | |
Collapse
|
50
|
Sandvig A, Berry M, Barrett LB, Butt A, Logan A. Myelin-, reactive glia-, and scar-derived CNS axon growth inhibitors: expression, receptor signaling, and correlation with axon regeneration. Glia 2004; 46:225-51. [PMID: 15048847 DOI: 10.1002/glia.10315] [Citation(s) in RCA: 285] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Axon regeneration is arrested in the injured central nervous system (CNS) by axon growth-inhibitory ligands expressed in oligodendrocytes/myelin, NG2-glia, and reactive astrocytes in the lesion and degenerating tracts, and by fibroblasts in scar tissue. Growth cone receptors (Rc) bind inhibitory ligands, activating a Rho-family GTPase intracellular signaling pathway that disrupts the actin cytoskeleton inducing growth cone collapse/repulsion. The known inhibitory ligands include the chondroitin sulfate proteoglycans (CSPG) Neurocan, Brevican, Phosphacan, Tenascin, and NG2, as either membrane-bound or secreted molecules; Ephrins expressed on astrocyte/fibroblast membranes; the myelin/oligodendrocyte-derived growth inhibitors Nogo, MAG, and OMgp; and membrane-bound semaphorins (Sema) produced by meningeal fibroblasts invading the scar. No definitive CSPG Rc have been identified, although intracellular signaling through the Rho family of G-proteins is probably common to all the inhibitory ligands. Ephrins bind to signalling Ephs. The ligand-binding Rc for all the myelin inhibitors is NgR and requires p75(NTR) for transmembrane signaling. The neuropilin (NP)/plexin (Plex) Rc complex binds Sema. Strategies for promoting axon growth after CNS injury are thwarted by the plethora of inhibitory ligands and the ligand promiscuity of some of their Rc. There is also paradoxical reciprocal expression of many of the inhibitory ligands/Rc in normal and damaged neurons, and NgR expression is restricted to a limited number of neuronal populations. All these factors, together with an incomplete understanding of the normal functions of many of these molecules in the intact CNS, presently confound interpretive acumen in regenerative studies.
Collapse
Affiliation(s)
- Axel Sandvig
- Laboratory of Regenerative Neurobiology, Institute for Experimental Medical Research, Ullevål University Hospital, Oslo, Norway.
| | | | | | | | | |
Collapse
|