1
|
Xue X, Wu X, Fan Y, Han S, Zhang H, Sun Y, Yin Y, Yin M, Chen B, Sun Z, Zhao S, Zhang Q, Liu W, Zhang J, Li J, Shi Y, Xiao Z, Dai J, Zhao Y. Heterogeneous fibroblasts contribute to fibrotic scar formation after spinal cord injury in mice and monkeys. Nat Commun 2024; 15:6321. [PMID: 39060269 PMCID: PMC11282111 DOI: 10.1038/s41467-024-50564-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Spinal cord injury (SCI) leads to fibrotic scar formation at the lesion site, yet the heterogeneity of fibrotic scar remains elusive. Here we show the heterogeneity in distribution, origin, and function of fibroblasts within fibrotic scars after SCI in mice and female monkeys. Utilizing lineage tracing and single-cell RNA sequencing (scRNA-seq), we found that perivascular fibroblasts (PFs), and meningeal fibroblasts (MFs), rather than pericytes/vascular smooth cells (vSMCs), primarily contribute to fibrotic scar in both transection and crush SCI. Crabp2 + /Emb+ fibroblasts (CE-F) derived from meninges primarily localize in the central region of fibrotic scars, demonstrating enhanced cholesterol synthesis and secretion of type I collagen and fibronectin. In contrast, perivascular/pial Lama1 + /Lama2+ fibroblasts (LA-F) are predominantly found at the periphery of the lesion, expressing laminin and type IV collagen and functionally involved in angiogenesis and lipid transport. These findings may provide a comprehensive understanding for remodeling heterogeneous fibrotic scars after SCI.
Collapse
Affiliation(s)
- Xiaoyu Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xianming Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuyu Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Haipeng Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuaijing Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaojiao Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiayin Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100101, China.
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
2
|
Holl D, Hau WF, Julien A, Banitalebi S, Kalkitsas J, Savant S, Llorens-Bobadilla E, Herault Y, Pavlovic G, Amiry-Moghaddam M, Dias DO, Göritz C. Distinct origin and region-dependent contribution of stromal fibroblasts to fibrosis following traumatic injury in mice. Nat Neurosci 2024; 27:1285-1298. [PMID: 38849523 PMCID: PMC11239523 DOI: 10.1038/s41593-024-01678-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/10/2024] [Indexed: 06/09/2024]
Abstract
Fibrotic scar tissue formation occurs in humans and mice. The fibrotic scar impairs tissue regeneration and functional recovery. However, the origin of scar-forming fibroblasts is unclear. Here, we show that stromal fibroblasts forming the fibrotic scar derive from two populations of perivascular cells after spinal cord injury (SCI) in adult mice of both sexes. We anatomically and transcriptionally identify the two cell populations as pericytes and perivascular fibroblasts. Fibroblasts and pericytes are enriched in the white and gray matter regions of the spinal cord, respectively. Both cell populations are recruited in response to SCI and inflammation. However, their contribution to fibrotic scar tissue depends on the location of the lesion. Upon injury, pericytes and perivascular fibroblasts become activated and transcriptionally converge on the generation of stromal myofibroblasts. Our results show that pericytes and perivascular fibroblasts contribute to the fibrotic scar in a region-dependent manner.
Collapse
Affiliation(s)
- Daniel Holl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wing Fung Hau
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Shatin, Hong Kong
| | - Anais Julien
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Shervin Banitalebi
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jannis Kalkitsas
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Soniya Savant
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Enric Llorens-Bobadilla
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Shatin, Hong Kong
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN-Institut Clinique de la Souris, Illkirch-Graffenstaden, France
| | - Guillaume Pavlovic
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN-Institut Clinique de la Souris, Illkirch-Graffenstaden, France
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - David Oliveira Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
- Center for Neuromusculoskeletal Restorative Medicine, Shatin, Hong Kong.
| |
Collapse
|
3
|
Bi Y, Duan W, Silver J. Collagen I is a critical organizer of scarring and CNS regeneration failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.592424. [PMID: 38766123 PMCID: PMC11100746 DOI: 10.1101/2024.05.07.592424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Although axotomized neurons retain the ability to initiate the formation of growth cones and attempt to regenerate after spinal cord injury, the scar area formed as a result of the lesion in most adult mammals contains a variety of reactive cells that elaborate multiple extracellular matrix and enzyme components that are not suitable for regrowth 1,2 . Newly migrating axons in the vicinity of the scar utilize upregulated LAR family receptor protein tyrosine phosphatases, such as PTPσ, to associate with extracellular chondroitin sulphate proteoglycans (CSPGs), which have been discovered to tightly entrap the regrowing axon tip and transform it into a dystrophic non-growing endball. The scar is comprised of two compartments, one in the lesion penumbra, the glial scar, composed of reactive microglia, astrocytes and OPCs; and the other in the lesion epicenter, the fibrotic scar, which is made up of fibroblasts, pericytes, endothelial cells and inflammatory cells. While the fibrotic scar is known to be strongly inhibitory, even more so than the glial scar, the molecular determinants that curtail axon elongation through the injury core are largely uncharacterized. Here, we show that one sole member of the entire family of collagens, collagen I, creates an especially potent inducer of endball formation and regeneration failure. The inhibitory signaling is mediated by mechanosensitive ion channels and RhoA activation. Staggered systemic administration of two blood-brain barrier permeable-FDA approved drugs, aspirin and pirfenidone, reduced fibroblast incursion into the complete lesion and dramatically decreased collagen I, as well as CSPG deposition which were accompanied by axonal growth and considerable functional recovery. The anatomical substrate for robust axonal regeneration was provided by laminin producing GFAP + and NG2 + bridging cells that spanned the wound. Our results reveal a collagen I-mechanotransduction axis that regulates axonal regrowth in spinal cord injury and raise a promising strategy for rapid clinical application.
Collapse
|
4
|
Guo J, Yang T, Zhang W, Yu K, Xu X, Li W, Song L, Gu X, Cao R, Cui S. Inhibition of CD44 suppresses the formation of fibrotic scar after spinal cord injury via the JAK2/STAT3 signaling pathway. iScience 2024; 27:108935. [PMID: 38323002 PMCID: PMC10846335 DOI: 10.1016/j.isci.2024.108935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/17/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Fibrotic scar is one of the main impediments to axon regeneration following spinal cord injury (SCI). In this study, we found that CD44 was upregulated during the formation of fibrotic scar, and blocking CD44 by IM7 caused downregulation of fibrosis-related extracellular matrix proteins at both 2 and 12 weeks post-spinal cord injury. More Biotinylated dextran amine (BDA)-traced corticospinal tract axons crossed the scar area and extended into the distal region after IM7 administration. A recovery of motor and sensory function was observed based on Basso Mouse Scale (BMS) scores and tail-flick test. In vitro experiments revealed that inhibiting CD44 and JAK2/STAT3 signaling pathway decreased the proliferation, differentiation, and migration of fibroblasts induced by the inflammatory supernatant. Collectively, these findings highlight the critical role of CD44 and its downstream JAK2/STAT3 signaling pathway in fibrotic scar formation, suggesting a potential therapeutic target for SCI.
Collapse
Affiliation(s)
- Jin Guo
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Tuo Yang
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Weizhong Zhang
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Kaiming Yu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Xiong Xu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Weizhen Li
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Lili Song
- Department of Hand & Microsurgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Rangjuan Cao
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Shusen Cui
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| |
Collapse
|
5
|
Abstract
Iron accumulation in the CNS occurs in many neurological disorders. It can contribute to neuropathology as iron is a redox-active metal that can generate free radicals. The reasons for the iron buildup in these conditions are varied and depend on which aspects of iron influx, efflux, or sequestration that help maintain iron homeostasis are dysregulated. Iron was shown recently to induce cell death and damage via lipid peroxidation under conditions in which there is deficient glutathione-dependent antioxidant defense. This form of cell death is called ferroptosis. Iron chelation has had limited success in the treatment of neurological disease. There is therefore much interest in ferroptosis as it potentially offers new drugs that could be more effective in reducing iron-mediated lipid peroxidation within the lipid-rich environment of the CNS. In this review, we focus on the molecular mechanisms that induce ferroptosis. We also address how iron enters and leaves the CNS, as well as the evidence for ferroptosis in several neurological disorders. Finally, we highlight biomarkers of ferroptosis and potential therapeutic strategies.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Fari Ryan
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Priya Jhelum
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Antje Kroner
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
6
|
Xu L, Nirwane A, Xu T, Kang M, Devasani K, Yao Y. Fibroblasts repair blood-brain barrier damage and hemorrhagic brain injury via TIMP2. Cell Rep 2022; 41:111709. [DOI: 10.1016/j.celrep.2022.111709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 09/11/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
|
7
|
Fibrosis in the central nervous system: from the meninges to the vasculature. Cell Tissue Res 2022; 387:351-360. [PMID: 34189605 PMCID: PMC8717837 DOI: 10.1007/s00441-021-03491-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023]
Abstract
Formation of a collagenous connective tissue scar after penetrating injuries to the brain or spinal cord has been described and investigated for well over 100 years. However, it was studied almost exclusively in the context of penetrating injuries that resulted in infiltration of meningeal fibroblasts, which raised doubts about translational applicability to most CNS injuries where the meninges remain intact. Recent studies demonstrating the perivascular niche as a source of fibroblasts have debunked the traditional view that a fibrotic scar only forms after penetrating lesions that tear the meninges. These studies have led to a renewed interest in CNS fibrosis not only in the context of axon regeneration after spinal cord injury, but also across a spectrum of CNS disorders. Arising with this renewed interest is some discrepancy about which perivascular cell gives rise to the fibrotic scar, but additional studies are beginning to provide some clarity. Although mechanistic studies on CNS fibrosis are still lacking, the similarities to fibrosis of other organs should provide important insight into how CNS fibrosis can be therapeutically targeted to promote functional recovery.
Collapse
|
8
|
Lee BJ, Jeong JH. Review: Steroid Use in Patients With Acute Spinal Cord Injury and Guideline Update. Korean J Neurotrauma 2022; 18:22-30. [PMID: 35557630 PMCID: PMC9064751 DOI: 10.13004/kjnt.2022.18.e21] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/05/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Byung-Jou Lee
- Department of Neurosurgery and Neuroscience & Radiosurgery Hybrid Research Center, Inje University Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Je Hoon Jeong
- Department of Neurosurgery, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| |
Collapse
|
9
|
Dorrier CE, Jones HE, Pintarić L, Siegenthaler JA, Daneman R. Emerging roles for CNS fibroblasts in health, injury and disease. Nat Rev Neurosci 2022; 23:23-34. [PMID: 34671105 PMCID: PMC8527980 DOI: 10.1038/s41583-021-00525-w] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2021] [Indexed: 02/06/2023]
Abstract
Recent transcriptomic, histological and functional studies have begun to shine light on the fibroblasts present in the meninges, choroid plexus and perivascular spaces of the brain and spinal cord. Although the origins and functions of CNS fibroblasts are still being described, it is clear that they represent a distinct cell population, or populations, that have likely been confused with other cell types on the basis of the expression of overlapping cellular markers. Recent work has revealed that fibroblasts play crucial roles in fibrotic scar formation in the CNS after injury and inflammation, which have also been attributed to other perivascular cell types such as pericytes and vascular smooth muscle cells. In this Review, we describe the current knowledge of the location and identity of CNS perivascular cell types, with a particular focus on CNS fibroblasts, including their origin, subtypes, roles in health and disease, and future areas for study.
Collapse
Affiliation(s)
- Cayce E Dorrier
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Hannah E Jones
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lucija Pintarić
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Julie A Siegenthaler
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Richard Daneman
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
10
|
Dorrier CE, Aran D, Haenelt EA, Sheehy RN, Hoi KK, Pintarić L, Chen Y, Lizama CO, Cautivo KM, Weiner GA, Popko B, Fancy SPJ, Arnold T, Daneman R. CNS fibroblasts form a fibrotic scar in response to immune cell infiltration. Nat Neurosci 2021; 24:234-244. [PMID: 33526922 PMCID: PMC7877789 DOI: 10.1038/s41593-020-00770-9] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Fibrosis is a common pathological response to inflammation in many peripheral tissues and can prevent tissue regeneration and repair. Here, we identified persistent fibrotic scarring in the CNS following immune cell infiltration in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Using lineage tracing and single-cell sequencing in EAE, we determined that the majority of the fibrotic scar is derived from proliferative CNS fibroblasts, not pericytes or infiltrating bone marrow-derived cells. Ablating proliferating fibrotic cells using cell-specific expression of herpes thymidine kinase led to an increase in oligodendrocyte lineage cells within the inflammatory lesions and a reduction in motor disability. We further identified that interferon-gamma pathway genes are enriched in CNS fibrotic cells, and the fibrotic cell-specific deletion of Ifngr1 resulted in reduced fibrotic scarring in EAE. These data delineate a framework for understanding the CNS fibrotic response.
Collapse
Affiliation(s)
- Cayce E. Dorrier
- Departments of Pharmacology and Neurosciences, University
of California San Diego, La Jolla, California 92093 USA
| | - Dvir Aran
- Technion- Israel Institute of Technology, Haifa,
Israel
| | - Ezekiel A. Haenelt
- Departments of Pharmacology and Neurosciences, University
of California San Diego, La Jolla, California 92093 USA
| | - Ryan N. Sheehy
- Departments of Pharmacology and Neurosciences, University
of California San Diego, La Jolla, California 92093 USA
| | - Kimberly K. Hoi
- Department of Neurology, University of California San
Francisco, San Francisco, California 94158 USA
| | - Lucija Pintarić
- Departments of Pharmacology and Neurosciences, University
of California San Diego, La Jolla, California 92093 USA
| | - Yanan Chen
- Department of Neurology, Feinberg School of Medicine,
Northwestern University, Chicago, Illinois 60611 USA
| | - Carlos O. Lizama
- Cardiovascular Research Institute, University of
California San Francisco, San Francisco, California 94158 USA
| | - Kelly M. Cautivo
- Department of Laboratory Medicine, University of
California San Francisco, San Francisco, California 94143 USA
| | - Geoffrey A. Weiner
- Departments of Pharmacology and Neurosciences, University
of California San Diego, La Jolla, California 92093 USA
| | - Brian Popko
- Department of Neurology, Feinberg School of Medicine,
Northwestern University, Chicago, Illinois 60611 USA
| | - Stephen P. J. Fancy
- Department of Neurology, University of California San
Francisco, San Francisco, California 94158 USA
| | - Thomas Arnold
- Department of Pediatrics, University of California San
Francisco, California 94143 USA.,Co-Corresponding authors: Thomas
Arnold, MD, , Richard Daneman, PhD,
| | - Richard Daneman
- Departments of Pharmacology and Neurosciences, University
of California San Diego, La Jolla, California 92093 USA.,Co-Corresponding authors: Thomas
Arnold, MD, , Richard Daneman, PhD,
| |
Collapse
|
11
|
Liu X, Liu Y, Jin H, Khodeiry MM, Kong W, Wang N, Lee JK, Lee RK. Reactive Fibroblasts in Response to Optic Nerve Crush Injury. Mol Neurobiol 2020; 58:1392-1403. [PMID: 33184784 DOI: 10.1007/s12035-020-02199-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/03/2020] [Indexed: 01/18/2023]
Abstract
Traumatic optic neuropathy leads to bidirectional degeneration of retinal ganglion cells and axons and results in optic nerve scaring, which inhibits the regeneration of damaged axons. Compared with its glial counterpart, the fibrotic response causing nerve scar tissue is poorly permissive to axonal regeneration. Using collagen1α1-GFP reporter mice, we characterize the development of fibrotic scar formation following optic nerve crush injury. We observe that perivascular collagen1α1 cells constitute a major cellular component of the fibrotic scar. We demonstrate that extracellular molecules and monocytes are key factors contributing to the pathogenesis of optic nerve fibrotic scar formation, with a previously unrecognized encapsulation of this scar. We also characterize the distribution of collagen1α1 cells in the retina after optic nerve crush injury based on in vivo and whole-mount retinal imaging. Our results identify collagen1α1 cells as a major component of fibrotic scarring following ONC and are a potential molecular target for promoting axonal regeneration after optic nerve injury.
Collapse
Affiliation(s)
- Xiangxiang Liu
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Eye Institute, Capital Medical University, Beijing, China
| | - Yuan Liu
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Huiyi Jin
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Mohamed M Khodeiry
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Department of Ophthalmology, Research Institute of Ophthalmology, Giza, 12557, Egypt
| | - Weizheng Kong
- School of Life Science, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Eye Institute, Capital Medical University, Beijing, China
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Richard K Lee
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
12
|
|
13
|
Bradbury EJ, Burnside ER. Moving beyond the glial scar for spinal cord repair. Nat Commun 2019; 10:3879. [PMID: 31462640 PMCID: PMC6713740 DOI: 10.1038/s41467-019-11707-7] [Citation(s) in RCA: 446] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 07/25/2019] [Indexed: 02/08/2023] Open
Abstract
Traumatic spinal cord injury results in severe and irreversible loss of function. The injury triggers a complex cascade of inflammatory and pathological processes, culminating in formation of a scar. While traditionally referred to as a glial scar, the spinal injury scar in fact comprises multiple cellular and extracellular components. This multidimensional nature should be considered when aiming to understand the role of scarring in limiting tissue repair and recovery. In this Review we discuss recent advances in understanding the composition and phenotypic characteristics of the spinal injury scar, the oversimplification of defining the scar in binary terms as good or bad, and the development of therapeutic approaches to target scar components to enable improved functional outcome after spinal cord injury.
Collapse
Affiliation(s)
- Elizabeth J Bradbury
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK.
| | - Emily R Burnside
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| |
Collapse
|
14
|
Stewart AN, Kendziorski G, Deak ZM, Bartosek NC, Rezmer BE, Jenrow K, Rossignol J, Dunbar GL. Transplantation of mesenchymal stem cells that overexpress NT-3 produce motor improvements without axonal regeneration following complete spinal cord transections in rats. Brain Res 2018; 1699:19-33. [DOI: 10.1016/j.brainres.2018.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/26/2018] [Accepted: 06/01/2018] [Indexed: 12/22/2022]
|
15
|
Abstract
A recent study indicates that reducing fibrotic scarring by genetically abrogating the proliferation of type A pericytes promotes axon regeneration and functional recovery after spinal cord injury. Questions remain regarding the identity of the cells being manipulated and the balance between the beneficial and detrimental effects of fibrotic scarring.
Collapse
|
16
|
Moreno PMD, Ferreira AR, Salvador D, Rodrigues MT, Torrado M, Carvalho ED, Tedebark U, Sousa MM, Amaral IF, Wengel J, Pêgo AP. Hydrogel-Assisted Antisense LNA Gapmer Delivery for In Situ Gene Silencing in Spinal Cord Injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:393-406. [PMID: 29858074 PMCID: PMC5992461 DOI: 10.1016/j.omtn.2018.03.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 03/04/2018] [Accepted: 03/15/2018] [Indexed: 12/16/2022]
Abstract
After spinal cord injury (SCI), nerve regeneration is severely hampered due to the establishment of a highly inhibitory microenvironment at the injury site, through the contribution of multiple factors. The potential of antisense oligonucleotides (AONs) to modify gene expression at different levels, allowing the regulation of cell survival and cell function, together with the availability of chemically modified nucleic acids with favorable biopharmaceutical properties, make AONs an attractive tool for novel SCI therapy developments. In this work, we explored the potential of locked nucleic acid (LNA)-modified AON gapmers in combination with a fibrin hydrogel bridging material to induce gene silencing in situ at a SCI lesion site. LNA gapmers were effectively developed against two promising gene targets aiming at enhancing axonal regeneration-RhoA and GSK3β. The fibrin-matrix-assisted AON delivery system mediated potent RNA knockdown in vitro in a dorsal root ganglion explant culture system and in vivo at a SCI lesion site, achieving around 75% downregulation 5 days after hydrogel injection. Our results show that local implantation of a AON-gapmer-loaded hydrogel matrix mediated efficient gene silencing in the lesioned spinal cord and is an innovative platform that can potentially combine gene regulation with regenerative permissive substrates aiming at SCI therapeutics and nerve regeneration.
Collapse
Affiliation(s)
- Pedro M D Moreno
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana R Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Daniela Salvador
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria T Rodrigues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Marília Torrado
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Eva D Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ulf Tedebark
- GE Healthcare Bio-Sciences AB, 75184 Uppsala, Sweden; SynMer AB, 17568 Järfälla, Sweden
| | - Mónica M Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Nerve Regeneration Group, Universidade do Porto, 4200-135 Porto, Portugal
| | - Isabel F Amaral
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Jesper Wengel
- Nucleic Acid Center, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark
| | - Ana P Pêgo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; Faculdade de Engenharia da Universidade do Porto, 4200-465 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
17
|
Dias DO, Kim H, Holl D, Werne Solnestam B, Lundeberg J, Carlén M, Göritz C, Frisén J. Reducing Pericyte-Derived Scarring Promotes Recovery after Spinal Cord Injury. Cell 2018; 173:153-165.e22. [PMID: 29502968 PMCID: PMC5871719 DOI: 10.1016/j.cell.2018.02.004] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/18/2017] [Accepted: 02/01/2018] [Indexed: 01/20/2023]
Abstract
CNS injury often severs axons. Scar tissue that forms locally at the lesion site is thought to block axonal regeneration, resulting in permanent functional deficits. We report that inhibiting the generation of progeny by a subclass of pericytes led to decreased fibrosis and extracellular matrix deposition after spinal cord injury in mice. Regeneration of raphespinal and corticospinal tract axons was enhanced and sensorimotor function recovery improved following spinal cord injury in animals with attenuated pericyte-derived scarring. Using optogenetic stimulation, we demonstrate that regenerated corticospinal tract axons integrated into the local spinal cord circuitry below the lesion site. The number of regenerated axons correlated with improved sensorimotor function recovery. In conclusion, attenuation of pericyte-derived fibrosis represents a promising therapeutic approach to facilitate recovery following CNS injury. Inhibition of pericyte proliferation reduces fibrotic scar tissue following injury Attenuated pericyte-derived scarring facilitates motor axon regeneration Regenerated axons functionally re-integrate into the local spinal circuitry Attenuated pericyte-derived scarring improves sensorimotor recovery
Collapse
Affiliation(s)
- David Oliveira Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Hoseok Kim
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Daniel Holl
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Beata Werne Solnestam
- Science for Life Laboratory, Karolinska Institutet Science Park, SE-171 65 Stockholm, Sweden
| | - Joakim Lundeberg
- Science for Life Laboratory, Karolinska Institutet Science Park, SE-171 65 Stockholm, Sweden
| | - Marie Carlén
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, SE-141 83 Huddinge, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
18
|
Non-functionalized soft alginate hydrogel promotes locomotor recovery after spinal cord injury in a rat hemimyelonectomy model. Acta Neurochir (Wien) 2018; 160:449-457. [PMID: 29230560 DOI: 10.1007/s00701-017-3389-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/31/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) and the consecutive devastating neurological sequelae have an enormous individual and economic impact. Implantation of functionalized hydrogels is a promising approach, because they can serve as a matrix for the regenerating tissue, carry and release bioactive molecules and various cell types. We already demonstrated that non-functionalized soft alginate hydrogel supported axonal outgrowth and protected neurons against oxidative stress in vitro. Here, we investigated the effects of such soft alginate hydrogels on locomotor recovery in small and large spinal cord lesions. METHOD Hemimyelonectomy of 2 mm or 4 mm length was performed in rats and soft alginate hydrogel was implanted. Functional recovery of the hindlimbs was assessed in the open field [Batto Beattie Bresnahan (BBB) score] and using swimming test [Louisville Swim score (LSS)] for 140 days post injury (DPI). Reference histology was performed. RESULTS Rats that received an alginate implant into 2 mm spinal cord lesions demonstrated significantly improved locomotor recovery compared to controls detectable already at 10 DPI. At 140 DPI, they reached higher LSS and BBB scores in swimming and open field tests, respectively. However, this beneficial effect of alginate was lacking in animals with larger (4 mm) lesions. Histological examination suggested that fibrous scarring in the spinal cord was reduced after alginate implantation in comparison to controls. CONCLUSIONS Implantation of soft alginate hydrogel in small spinal cord lesions improved functional recovery. Possible underlying mechanisms include the mechanical stabilization of the wound, reduction of secondary damage and inhibition of fibrous scarring.
Collapse
|
19
|
Dias DO, Göritz C. Fibrotic scarring following lesions to the central nervous system. Matrix Biol 2018; 68-69:561-570. [PMID: 29428230 DOI: 10.1016/j.matbio.2018.02.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 10/18/2022]
Abstract
Following lesions to the central nervous system, scar tissue forms at the lesion site. Injury often severs axons and scar tissue is thought to block axonal regeneration, resulting in permanent functional deficits. While scar-forming astrocytes have been extensively studied, much less attention has been given to the fibrotic, non-glial component of the scar. We here review recent progress in understanding fibrotic scar formation following different lesions to the brain and spinal cord. We specifically highlight recent evidence for pericyte-derived fibrotic scar tissue formation, discussing the origin, recruitment, function and therapeutic relevance of fibrotic scarring.
Collapse
Affiliation(s)
- David Oliveira Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
20
|
Ahuja CS, Nori S, Tetreault L, Wilson J, Kwon B, Harrop J, Choi D, Fehlings MG. Traumatic Spinal Cord Injury-Repair and Regeneration. Neurosurgery 2017; 80:S9-S22. [PMID: 28350947 DOI: 10.1093/neuros/nyw080] [Citation(s) in RCA: 561] [Impact Index Per Article: 70.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/12/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Traumatic spinal cord injuries (SCI) have devastating consequences for the physical, financial, and psychosocial well-being of patients and their caregivers. Expediently delivering interventions during the early postinjury period can have a tremendous impact on long-term functional recovery. PATHOPHYSIOLOGY This is largely due to the unique pathophysiology of SCI where the initial traumatic insult (primary injury) is followed by a progressive secondary injury cascade characterized by ischemia, proapoptotic signaling, and peripheral inflammatory cell infiltration. Over the subsequent hours, release of proinflammatory cytokines and cytotoxic debris (DNA, ATP, reactive oxygen species) cyclically adds to the harsh postinjury microenvironment. As the lesions mature into the chronic phase, regeneration is severely impeded by the development of an astroglial-fibrous scar surrounding coalesced cystic cavities. Addressing these challenges forms the basis of current and upcoming treatments for SCI. MANAGEMENT This paper discusses the evidence-based management of a patient with SCI while emphasizing the importance of early definitive care. Key neuroprotective therapies are summarized including surgical decompression, methylprednisolone, and blood pressure augmentation. We then review exciting neuroprotective interventions on the cusp of translation such as Riluzole, Minocycline, magnesium, therapeutic hypothermia, and CSF drainage. We also explore the most promising neuroregenerative strategies in trial today including Cethrin™, anti-NOGO antibody, cell-based approaches, and bioengineered biomaterials. Each section provides a working knowledge of the key preclinical and patient trials relevant to clinicians while highlighting the pathophysiologic rationale for the therapies. CONCLUSION We conclude with our perspectives on the future of treatment and research in this rapidly evolving field.
Collapse
Affiliation(s)
- Christopher S Ahuja
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,Department of Genetics and Development, University of Toronto, Toronto, Canada
| | - Satoshi Nori
- Department of Genetics and Development, University of Toronto, Toronto, Canada
| | | | - Jefferson Wilson
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,Spine Program, University of Toronto, Toronto, Canada
| | - Brian Kwon
- Vancouver Spine Institute, Vancouver General Hospital, Vancouver, Canada.,Department of Surgery, University of British Columbia, Vancouver, Canada
| | - James Harrop
- Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - David Choi
- National Hospital for Neurology and Neurosurgery, University College London, London, England
| | - Michael G Fehlings
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,Spine Program, University of Toronto, Toronto, Canada.,Department of Genetics and Development, University of Toronto, Toronto, Canada
| |
Collapse
|
21
|
Li D, Tong L, Kawano H, Liu N, Liu L, Li HP. Protective effects of batroxobin on a nigrostriatal pathway injury in mice. Brain Res Bull 2016; 127:195-201. [PMID: 27679398 DOI: 10.1016/j.brainresbull.2016.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury triggers a series of damaged processes, such as neuronal death and apoptosis, inflammation and scar formation, which contribute to evolution of brain injury. The present study investigated the neuroprotective effects of batroxobin, a drug widely used clinically for ischemia, in a nigrostriatal pathway injury model. Mice subjected to the nigrostriatal pathway injury were injected with batroxobin (30 BU/kg) or vehicle immediately after injury. The behavioral studies showed that batroxobin could improve the motor function in injured mice in long term. Batroxobin also reduced neuronal apoptosis and inflammation at the acute stage. Moreover, administration of batroxobin attenuated the scar formation and reduced the lesion size at 4 and 14days after brain injury. These results suggest that batroxobin has beneficial effects on the nigrostriatal pathway injury, indicating a potential clinical application.
Collapse
Affiliation(s)
- Dan Li
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lei Tong
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hitoshi Kawano
- Department of Health and Dietetics, Faculty of Health and Medical Science, Teikyo Heisei University, Tokyo 170-8445, Japan
| | - Nan Liu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lu Liu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hong-Peng Li
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
22
|
Vangansewinkel T, Geurts N, Quanten K, Nelissen S, Lemmens S, Geboes L, Dooley D, Vidal PM, Pejler G, Hendrix S. Mast cells promote scar remodeling and functional recovery after spinal cord injury via mouse mast cell protease 6. FASEB J 2016; 30:2040-57. [PMID: 26917739 DOI: 10.1096/fj.201500114r] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/28/2016] [Indexed: 12/12/2022]
Abstract
An important barrier for axon regeneration and recovery after traumatic spinal cord injury (SCI) is attributed to the scar that is formed at the lesion site. Here, we investigated the effect of mouse mast cell protease (mMCP) 6, a mast cell (MC)-specific tryptase, on scarring and functional recovery after a spinal cord hemisection injury. Functional recovery was significantly impaired in both MC-deficient and mMCP6-knockout (mMCP6(-/-)) mice after SCI compared with wild-type control mice. This decrease in locomotor performance was associated with an increased lesion size and excessive scarring at the injury site. Axon growth-inhibitory chondroitin sulfate proteoglycans and the extracellular matrix components fibronectin, laminin, and collagen IV were significantly up-regulated in MC-deficient and mMCP6(-/-) mice, with an increase in scar volume between 23 and 32%. A degradation assay revealed that mMCP6 directly cleaves fibronectin and collagen IV in vitro In addition, gene expression levels of the scar components fibronectin, aggrecan, and collagen IV were increased up to 6.8-fold in mMCP6(-/-) mice in the subacute phase after injury. These data indicate that endogenous mMCP6 has scar-suppressing properties after SCI via indirect cleavage of axon growth-inhibitory scar components and alteration of the gene expression profile of these factors.-Vangansewinkel, T., Geurts, N., Quanten, K., Nelissen, S., Lemmens, S., Geboes, L., Dooley, D., Vidal, P. M., Pejler, G., Hendrix, S. Mast cells promote scar remodeling and functional recovery after spinal cord injury via mouse mast cell protease 6.
Collapse
Affiliation(s)
- Tim Vangansewinkel
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Nathalie Geurts
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Kirsten Quanten
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sofie Nelissen
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Stefanie Lemmens
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Lies Geboes
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Dearbhaile Dooley
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Pia M Vidal
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Gunnar Pejler
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden; and Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium;
| |
Collapse
|
23
|
Pharmacological Suppression of CNS Scarring by Deferoxamine Reduces Lesion Volume and Increases Regeneration in an In Vitro Model for Astroglial-Fibrotic Scarring and in Rat Spinal Cord Injury In Vivo. PLoS One 2015. [PMID: 26222542 PMCID: PMC4519270 DOI: 10.1371/journal.pone.0134371] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lesion-induced scarring is a major impediment for regeneration of injured axons in the central nervous system (CNS). The collagen-rich glial-fibrous scar contains numerous axon growth inhibitory factors forming a regeneration-barrier for axons. We demonstrated previously that the combination of the iron chelator 2,2’-bipyridine-5,5’-decarboxylic acid (BPY-DCA) and 8-Br-cyclic AMP (cAMP) inhibits scar formation and collagen deposition, leading to enhanced axon regeneration and partial functional recovery after spinal cord injury. While BPY-DCA is not a clinical drug, the clinically approved iron chelator deferoxamine mesylate (DFO) may be a suitable alternative for anti-scarring treatment (AST). In order to prove the scar-suppressing efficacy of DFO we modified a recently published in vitro model for CNS scarring. The model comprises a co-culture system of cerebral astrocytes and meningeal fibroblasts, which form scar-like clusters when stimulated with transforming growth factor-β (TGF-β). We studied the mechanisms of TGF-β-induced CNS scarring and compared the efficiency of different putative pharmacological scar-reducing treatments, including BPY-DCA, DFO and cAMP as well as combinations thereof. We observed modulation of TGF-β-induced scarring at the level of fibroblast proliferation and contraction as well as specific changes in the expression of extracellular matrix molecules and axon growth inhibitory proteins. The individual and combinatorial pharmacological treatments had distinct effects on the cellular and molecular aspects of in vitro scarring. DFO could be identified as a putative anti-scarring treatment for CNS trauma. We subsequently validated this by local application of DFO to a dorsal hemisection in the rat thoracic spinal cord. DFO treatment led to significant reduction of scarring, slightly increased regeneration of corticospinal tract as well as ascending CGRP-positive axons and moderately improved locomotion. We conclude that the in vitro model for CNS scarring is suitable for efficient pre-screening and identification of putative scar-suppressing agents prior to in vivo application and validation, thus saving costs, time and laboratory animals.
Collapse
|
24
|
Hong J, Nandiwada V, Jones V, Lu M, Warner DS, Mukhopadhyay S, Sheng H. CB1 cannabinoid receptor agonist inhibits matrix metalloproteinase activity in spinal cord injury: A possible mechanism of improved recovery. Neurosci Lett 2015; 597:19-24. [DOI: 10.1016/j.neulet.2015.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 02/19/2015] [Accepted: 04/09/2015] [Indexed: 10/23/2022]
|
25
|
A Fab fragment directed against the neural cell adhesion molecule L1 enhances functional recovery after injury of the adult mouse spinal cord. Biochem J 2014; 460:437-46. [DOI: 10.1042/bj20131677] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A recombinant monovalent Fab fragment recognizing a functional epitope within the third fibronectin type III domain of murine cell adhesion molecule L1 induces neurite outgrowth and neuronal survival in vitro and enhances functional recovery after spinal cord injury in mice.
Collapse
|
26
|
Schreiber J, Schachner M, Schumacher U, Lorke DE. Extracellular matrix alterations, accelerated leukocyte infiltration and enhanced axonal sprouting after spinal cord hemisection in tenascin-C-deficient mice. Acta Histochem 2013; 115:865-78. [PMID: 23701962 DOI: 10.1016/j.acthis.2013.04.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 01/08/2023]
Abstract
The extracellular matrix glycoprotein tenascin-C has been implicated in wound repair and axonal growth. Its role in mammalian spinal cord injury is largely unknown. In vitro it can be both neurite-outgrowth promoting and repellent. To assess its effects on glial reactions, extracellular matrix formation, and axonal regrowth/sprouting in vivo, 20 tenascin-C-deficient and 20 wild type control mice underwent lumbar spinal cord hemisection. One, three, seven and fourteen days post-surgery, cryostat sections of the spinal cord were examined by conventional histology and by immunohistochemistry using antibodies against F4/80 (microglia/macrophage), GFAP (astroglia), neurofilament, fibronectin, laminin and collagen type IV. Fibronectin immunoreactivity was significantly down-regulated in tenascin-C-deficient mice. Moreover, fourteen days after injury, immunodensity of neurofilament-positive fibers was two orders of magnitude higher along the incision edges of tenascin-C-deficient mice as compared to control mice. In addition, lymphocyte infiltration was seen two days earlier in tenascin-C-deficient mice than in control mice and neutrophil infiltration was increased seven days after injury. The increase in thin neurofilament positive fibers in tenascin-C-deficient mice indicates that lack of tenascin-C alters the inflammatory reaction and extracellular matrix composition in a way that penetration of axonal fibers into spinal cord scar tissue may be facilitated.
Collapse
Affiliation(s)
- Jenny Schreiber
- University Medical Center Hamburg-Eppendorf, Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, Martinistraße 52, 20246 Hamburg, Germany
| | | | | | | |
Collapse
|
27
|
Austin JW, Afshar M, Fehlings MG. The relationship between localized subarachnoid inflammation and parenchymal pathophysiology after spinal cord injury. J Neurotrauma 2013; 29:1838-49. [PMID: 22655536 DOI: 10.1089/neu.2012.2354] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Subarachnoid inflammation following spinal cord injury (SCI) can lead to the formation of localized subarachnoid scarring and the development of post-traumatic syringomyelia (PTS). While PTS is a devastating complication of SCI, its relative rarity (occurring symptomatically in about 5% of clinical cases), and lack of fundamental physiological insights, have led us to examine an animal model of traumatic SCI with induced arachnoiditis. We hypothesized that arachnoiditis associated with SCI would potentiate early parenchymal pathophysiology. To test this theory, we examined early spatial pathophysiology in four groups: (1) sham (non-injured controls), (2) arachnoiditis (intrathecal injection of kaolin), (3) SCI (35-g clip contusion/compression injury), and (4) PTS (intrathecal kaolin+SCI). Overall, there was greater parenchymal inflammation and scarring in the PTS group relative to the SCI group. This was demonstrated by significant increases in cytokine (IL-1α and IL-1β) and chemokine (MCP-1, GRO/KC, and MIP-1α) production, MPO activity, blood-spinal cord barrier (BSCB) permeability, and MMP-9 activity. However, parenchymal inflammatory mediator production (acute IL-1α and IL-1β, subacute chemokines), BSCB permeability, and fibrous scarring in the PTS group were larger than the sum of the SCI group and arachnoiditis group combined, suggesting that arachnoiditis does indeed potentiate parenchymal pathophysiology. Accordingly, these findings suggest that the development of arachnoiditis associated with SCI can lead to an exacerbation of the parenchymal injury, potentially impacting the outcome of this devastating condition.
Collapse
Affiliation(s)
- James W Austin
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
28
|
Blomster LV, Cowin GJ, Kurniawan ND, Ruitenberg MJ. Detection of endogenous iron deposits in the injured mouse spinal cord through high-resolution ex vivo and in vivo MRI. NMR IN BIOMEDICINE 2013; 26:141-150. [PMID: 22730180 DOI: 10.1002/nbm.2829] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 05/07/2012] [Accepted: 05/25/2012] [Indexed: 06/01/2023]
Abstract
The main aim of this study was to employ high-resolution MRI to investigate the spatiotemporal development of pathological features associated with contusive spinal cord injury (SCI) in mice. Experimental mice were subjected to either sham surgery or moderate contusive SCI. A 16.4-T small-animal MR system was employed for nondestructive imaging of post-mortem, fixed spinal cord specimens at the subacute (7 days) and more chronic (28-35 days) stages post-injury. Routine histological techniques were used for subsequent investigation of the observed neuropathology at the microscopic level. The central core of the lesion appeared as a dark hypo-intense area on MR images at all time points investigated. Small focal hypo-intense spots were also observed spreading through the dorsal funiculi proximal and distal to the site of impact, an area that is known to undergo gliosis and Wallerian degeneration in response to injury. Histological examination revealed these hypo-intense spots to be high in iron content as determined by Prussian blue staining. Quantitative image analysis confirmed the increased presence of iron deposits at all post-injury time points investigated (p<0.05). Distant iron deposits were also detectable through live imaging without the use of contrast-enhancing agents, enabling the longitudinal investigation of this pathology in individual animals. Further immunohistochemical evaluation showed that intracellular iron deposits localised to macrophages/microglia, astrocytes and oligodendrocytes in the subacute phase of SCI, but predominantly to glial fibrillary acidic protein-positive, CC-1-positive astrocytes at later stages of recovery. Progressive, widespread intracellular iron accumulation is thus a normal feature of SCI in mice, and high-resolution MRI can be effectively used to detect and monitor these neuropathological changes with time.
Collapse
Affiliation(s)
- Linda V Blomster
- University of Queensland, School of Biomedical Sciences, St Lucia, Qld, Australia
| | | | | | | |
Collapse
|
29
|
Advances in natural biomaterials for nerve tissue repair. Neurosci Lett 2012; 519:103-14. [DOI: 10.1016/j.neulet.2012.02.027] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 02/06/2012] [Accepted: 02/08/2012] [Indexed: 12/22/2022]
|
30
|
Kruse F, Bosse F, Vogelaar CF, Brazda N, Küry P, Gasis M, Müller HW. Cortical gene expression in spinal cord injury and repair: insight into the functional complexity of the neural regeneration program. Front Mol Neurosci 2011; 4:26. [PMID: 21994489 PMCID: PMC3182759 DOI: 10.3389/fnmol.2011.00026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/05/2011] [Indexed: 11/29/2022] Open
Abstract
Traumatic spinal cord injury (SCI) results in the formation of a fibrous scar acting as a growth barrier for regenerating axons at the lesion site. We have previously shown (Klapka et al., 2005) that transient suppression of the inhibitory lesion scar in rat spinal cord leads to long distance axon regeneration, retrograde rescue of axotomized cortical motoneurons, and improvement of locomotor function. Here we applied a systemic approach to investigate for the first time specific and dynamic alterations in the cortical gene expression profile following both thoracic SCI and regeneration-promoting anti-scarring treatment (AST). In order to monitor cortical gene expression we carried out microarray analyses using total RNA isolated from layer V/VI of rat sensorimotor cortex at 1–60 days post-operation (dpo). We demonstrate that cortical neurons respond to injury by massive changes in gene expression, starting as early as 1 dpo. AST, in turn, results in profound modifications of the lesion-induced expression profile. The treatment attenuates SCI-triggered transcriptional changes of genes related to inhibition of axon growth and impairment of cell survival, while upregulating the expression of genes associated with axon outgrowth, cell protection, and neural development. Thus, AST not only modifies the local environment impeding spinal cord regeneration by reduction of fibrous scarring in the injured spinal cord, but, in addition, strikingly changes the intrinsic capacity of cortical pyramidal neurons toward enhanced cell maintenance and axonal regeneration.
Collapse
Affiliation(s)
- Fabian Kruse
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
31
|
Jaerve A, Schiwy N, Schmitz C, Mueller HW. Differential effect of aging on axon sprouting and regenerative growth in spinal cord injury. Exp Neurol 2011; 231:284-94. [PMID: 21806987 DOI: 10.1016/j.expneurol.2011.07.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 07/08/2011] [Accepted: 07/14/2011] [Indexed: 12/22/2022]
Abstract
The demographics of acute spinal cord injury (SCI) are changing with an increased incidence in older age. However, the influence of aging on the regenerative growth potential of central nervous system (CNS) axons following SCI is not known. We investigated axonal sprouting along with the efficiency of the infusion of the stromal cell-derived growth factor-1 (SDF-1/CXCL12) and regenerative growth along with the anti-scarring treatment (AST) in young (2-3 months) and geriatric (22-28 months) female rats following SCI. AST included local injection of iron chelator (2,2'-dipyridine-5,5'-dicarboxylic acid) and 8-bromo-cyclic adenosine monophosphate solution into the lesion core. Axon outgrowth was investigated by immunohistological methods at 5 weeks after a partial dorsal hemisection at thoracic level T8. We found that aging significantly reduces spontaneous axon sprouting of corticospinal (CST), serotonergic (5-HT) raphespinal and catecholaminergic (TH) coerulospinal tracts in distinct regions of the spinal cord rostral to the lesion. However, impairment of axon sprouting could be markedly attenuated in geriatric animals by local infusion of SDF-1. Unexpectedly and in contrast to rostral sprouting, aging does not diminish the regenerative growth capacity of 5-HT-, TH- and calcitonin gene-related peptide (CGRP)-immunoreactive axons at 5 weeks after SCI. Moreover, 5-HT and TH axons maintain the ability to react upon AST with significantly enhanced regeneration in aged animals. These data are the first to demonstrate, that old age compromises axonal plasticity, but not regenerative growth, after SCI in a fiber tract-specific manner. Furthermore, AST and SDF-1 infusions remain efficient, which implicates that therapy in elderly patients is still feasible.
Collapse
Affiliation(s)
- Anne Jaerve
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | | | | | | |
Collapse
|
32
|
Gueye Y, Ferhat L, Sbai O, Bianco J, Ould-Yahoui A, Bernard A, Charrat E, Chauvin JP, Risso JJ, Féron F, Rivera S, Khrestchatisky M. Trafficking and secretion of matrix metalloproteinase-2 in olfactory ensheathing glial cells: A role in cell migration? Glia 2011; 59:750-70. [DOI: 10.1002/glia.21146] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 12/20/2010] [Indexed: 02/06/2023]
|
33
|
Yoshioka N, Kimura-Kuroda J, Saito T, Kawamura K, Hisanaga SI, Kawano H. Small molecule inhibitor of type I transforming growth factor-β receptor kinase ameliorates the inhibitory milieu in injured brain and promotes regeneration of nigrostriatal dopaminergic axons. J Neurosci Res 2010; 89:381-93. [PMID: 21259325 DOI: 10.1002/jnr.22552] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 10/14/2010] [Accepted: 10/19/2010] [Indexed: 12/15/2022]
Abstract
Transforming growth factor-β (TGF-β), a multifunctional cytokine, plays a crucial role in wound healing in the damaged central nervous system. To examine effects of the TGF-β signaling inhibition on formation of scar tissue and axonal regeneration, the small molecule inhibitor of type I TGF-β receptor kinase LY-364947 was continuously infused in the lesion site of mouse brain after a unilateral transection of the nigrostriatal dopaminergic pathway. At 2 weeks after injury, the fibrotic scar comprising extracellular matrix molecules including fibronectin, type IV collagen, and chondroitin sulfate proteoglycans was formed in the lesion center, and reactive astrocytes were increased around the fibrotic scar. In the brain injured and infused with LY-364947, fibrotic scar formation was suppressed and decreased numbers of reactive astrocytes occupied the lesion site. Although leukocytes and serum IgG were observed within the fibrotic scar in the injured brain, they were almost absent in the injured and LY-364947-treated brain. At 2 weeks after injury, tyrosine hydroxylase (TH)-immunoreactive fibers barely extended beyond the fibrotic scar in the injured brain, but numerous TH-immunoreactive fibers regenerated over the lesion site in the LY-364947-treated brain. These results indicate that inhibition of TGF-β signaling suppresses formation of the fibrotic scar and creates a permissive environment for axonal regeneration.
Collapse
Affiliation(s)
- Nozomu Yoshioka
- Department of Developmental Morphology, Tokyo Metropolitan Institute for Neuroscience, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Yoshioka N, Hisanaga SI, Kawano H. Suppression of fibrotic scar formation promotes axonal regeneration without disturbing blood-brain barrier repair and withdrawal of leukocytes after traumatic brain injury. J Comp Neurol 2010; 518:3867-81. [PMID: 20653039 DOI: 10.1002/cne.22431] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The fibrotic scar containing type IV collagen (Col IV) formed in a lesion site is considered as an obstacle to axonal regeneration, because intracerebral injection of 2,2'-dipyridyl (DPY), an inhibitor of Col IV triple-helix formation, suppresses fibrotic scar formation in the lesion site and promotes axonal regeneration. To determine the role of the fibrotic scar on the healing process of injured central nervous system (CNS), the restoration of blood-brain barrier (BBB) and withdrawal of inflammatory leukocytes were examined in mice subjected to unilateral transection of the nigrostriatal dopaminergic pathway and intracerebral DPY injection. At 5 days after injury, destruction of BBB represented by leakage of Evans blue (EB) and widespread infiltration of CD45-immunoreactive leukocytes was observed around the lesion site, whereas reactive astrocytes increased surrounding the BBB-destroyed area. By 2 weeks after injury, the region of EB leakage and the diffusion of leukocytes were restricted to the inside of the fibrotic scar, and reactive astrocytes gathered around the fibrotic scar. In the DPY-treated lesion site, formation of the fibrotic scar was suppressed (84% decrease in Col IV-deposited area), reactive astrocytes occupied the lesion center, and areas of both EB leakage and leukocyte infiltration decreased by 86%. DPY treatment increased the number of regenerated dopaminergic axons by 2.53-fold. These results indicate that suppression of fibrotic scar formation does not disturb the healing process in damaged CNS, and suggest that this strategy is a reliable tool to promote axonal regeneration after traumatic injury in the CNS.
Collapse
Affiliation(s)
- Nozomu Yoshioka
- Department of Developmental Morphology, Tokyo Metropolitan Institute for Neuroscience, Fuchu, Tokyo 183-8526, Japan
| | | | | |
Collapse
|
35
|
Affiliation(s)
- Michael G. Fehlings
- 1Division of Genetics and Development, Toronto Western Research Institute
- 2Institute of Medical Science, and
- 3Division of Neurosurgery, University of Toronto, Ontario, Canada
- 4Neural Repair and Regeneration, Spinal Program, University Health Network; and
| | - Gregory W. J. Hawryluk
- 1Division of Genetics and Development, Toronto Western Research Institute
- 2Institute of Medical Science, and
- 3Division of Neurosurgery, University of Toronto, Ontario, Canada
| |
Collapse
|
36
|
Marx SC, Kumar P, Dhalapathy S, Anitha Marx C. A comparative microanatomical study on cross sections of medial and lateral cutaneous nerves of forearm at the antecubital fossa: a cadaveric study. Ann Anat 2010; 192:107-15. [PMID: 20138488 DOI: 10.1016/j.aanat.2009.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 12/15/2009] [Accepted: 12/16/2009] [Indexed: 10/19/2022]
Abstract
BACKGROUND The anterior branch of the medial antebrachial cutaneous nerve of the forearm (AMACN) and the lateral antebrachial cutaneous nerve of the forearm (LACN) are used as potential donor grafts for repairing sensory nerves. A higher percentage of connective tissue plays an important role in predicting prognosis after nerve repair. The aim is to perform a comparative study on cross-sectional microanatomy and age related changes in non-fascicular components of the AMACN and LACN. METHODS Thirty six fresh human (from both sides of 14 male and 4 female) cadaveric AMACN and LACN were collected at antecubital fossae and studied at different magnifications for morphometric analysis (total cross-sectional area (Asc), fascicular area (Af) and non-fascicular area (Anonf)), after histological (Masson's trichrome stain) processing. RESULTS AMACN and LACN belong to polyfascicular type and showed differences in amount of connective and adipose tissues in non-fascicular areas. In the AMACN, there was less adipose tissue (19.38% in Asc and 25.57% in Anonf) with more collagen fibers (57.28% in Asc and 75.57% in Anonf) and in the LACN, there was more adipose tissue (47.51% in Asc and 58.19% in Anonf) with fewer collagen fibers (34.10% in Asc and 41.76% in Anonf) in interfascicular domains. CONCLUSIONS The amount of adipose tissue in LACN non-fascicular area was found to be high at all ages. The presence of less adipose tissue and collagen fibers in the non-fascicular area of the AMACN (below 60 years) could be used for successful nerve grafting when compared to LACN.
Collapse
Affiliation(s)
- S Chakravarthy Marx
- Department of Anatomy, Kasturba Medical College, Madhav nagar, Manipal 576104, Karnataka, India.
| | | | | | | |
Collapse
|
37
|
Leach JB, Achyuta AKH, Murthy SK. Bridging the Divide between Neuroprosthetic Design, Tissue Engineering and Neurobiology. FRONTIERS IN NEUROENGINEERING 2010; 2:18. [PMID: 20161810 PMCID: PMC2821180 DOI: 10.3389/neuro.16.018.2009] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 12/28/2009] [Indexed: 11/19/2022]
Abstract
Neuroprosthetic devices have made a major impact in the treatment of a variety of disorders such as paralysis and stroke. However, a major impediment in the advancement of this technology is the challenge of maintaining device performance during chronic implantation (months to years) due to complex intrinsic host responses such as gliosis or glial scarring. The objective of this review is to bring together research communities in neurobiology, tissue engineering, and neuroprosthetics to address the major obstacles encountered in the translation of neuroprosthetics technology into long-term clinical use. This article draws connections between specific challenges faced by current neuroprosthetics technology and recent advances in the areas of nerve tissue engineering and neurobiology. Within the context of the device-nervous system interface and central nervous system implants, areas of synergistic opportunity are discussed, including platforms to present cells with multiple cues, controlled delivery of bioactive factors, three-dimensional constructs and in vitro models of gliosis and brain injury, nerve regeneration strategies, and neural stem/progenitor cell biology. Finally, recent insights gained from the fields of developmental neurobiology and cancer biology are discussed as examples of exciting new biological knowledge that may provide fresh inspiration toward novel technologies to address the complexities associated with long-term neuroprosthetic device performance.
Collapse
Affiliation(s)
- Jennie B. Leach
- Department of Chemical and Biochemical Engineering, University of MarylandBaltimore, MD, USA
| | | | - Shashi K. Murthy
- Department of Chemical Engineering, Northeastern UniversityBoston, MA, USA
| |
Collapse
|