1
|
Leite C, Russo T, Cuccaro A, Pinto J, Polese G, Soares AM, Pretti C, Pereira E, Freitas R. The role of warming in modulating neodymium effects on adults and sperm of Mytilus galloprovincialis. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 358:120854. [PMID: 38640759 DOI: 10.1016/j.jenvman.2024.120854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/21/2024]
Abstract
The use of rare earth elements (REEs) has been increasing and one of the most used is neodymium (Nd). Being an emergent contaminant, its negative impacts are poorly understood. Aquatic organisms are also threatened by climate change-related factors, as is the case of warming, which can change the effects of REEs. Thus, the impacts of Nd, warming, and the combination of both stressors were studied in adult mussels and sperm of the species Mytilus galloprovincialis, after an exposure period of 28 days (adults) and 30 min (sperm). The effects were evaluated through the analysis of biochemical and histopathological alterations in adults and biochemical and physiological responses given by sperm. The results showed that mussels only activated their biotransformation capacity when exposed to the stressors acting alone, which was insufficient to avoid lipid peroxidation. Furthermore, warming (alone and combined with Nd) also produces damage to proteins. The digestive gland was the most sensitive organ to Nd, presenting several histopathological alterations. In the case of sperm, all stressors induced lipid peroxidation, a higher oxygen demand, and a decrease in velocity, even if the sperm viability was maintained. It seems that warming influenced the effects of Nd to some extent. The present findings contribute significantly to the field of REEs environmental toxicology by offering valuable insights into the impacts of Nd on various biological levels of mussels. Additionally, within the context of climate change, this study sheds light on how temperature influences the effects of Nd. The obtained results indicate that both stressors can potentially compromise the overall health of mussel populations, thereby affecting other species reliant on them for food and habitat. Moreover, this study highlights impaired sperm health, which could adversely affect their reproductive capacity and ultimately lead to population decline.
Collapse
Affiliation(s)
- Carla Leite
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Tania Russo
- Department of Biology, University of Naples Federico II, 80126, Napoli, Italy
| | - Alessia Cuccaro
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal; Department of Veterinary Sciences, University of Pisa, San Piero a Grado, 56122, Pisa, Italy
| | - João Pinto
- Department of Chemistry & LAQV-REQUIMTE, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Gianluca Polese
- Department of Biology, University of Naples Federico II, 80126, Napoli, Italy
| | - Amadeu Mvm Soares
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Carlo Pretti
- Department of Veterinary Sciences, University of Pisa, San Piero a Grado, 56122, Pisa, Italy; Interuniversity Consortium of Marine Biology of Leghorn "G. Bacci", 57128, Livorno, Italy
| | - Eduarda Pereira
- Department of Chemistry & LAQV-REQUIMTE, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rosa Freitas
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
2
|
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
|
3
|
Manesco C, Saavedra-Villanueva O, Martin M, de Lizaraga J, Varga B, Cloitre T, Gerber YN, Perrin FE, Gergely C. Organization of collagen fibers and tissue hardening: Markers of fibrotic scarring after spinal cord injury in mice revealed by multiphoton-atomic force microscopy imaging. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 53:102699. [PMID: 37572769 DOI: 10.1016/j.nano.2023.102699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/14/2023]
Abstract
Spinal cord injury is a dramatic disease leading to severe motor, sensitive and autonomic impairments. After injury the axonal regeneration is partly inhibited by the glial scar, acting as a physical and chemical barrier. The scarring process involves microglia, astrocytes and extracellular matrix components, such as collagen, constructing the fibrotic component of the scar. To investigate the role of collagen, we used a multimodal label-free imaging approach combining multiphoton and atomic force microscopy. The second harmonic generation signal exhibited by fibrillar collagen enabled to specifically monitor it as a biomarker of the lesion. An increase in collagen density and the formation of more tortuous fibers over time after injury are observed. Nano-mechanical investigations revealed a noticeable hardening of the injured area, correlated with collagen fibers' formation. These observations indicate the concomitance of important structural and mechanical modifications during the fibrotic scar evolution.
Collapse
Affiliation(s)
| | | | - Marta Martin
- L2C, Univ Montpellier, CNRS, Montpellier, France
| | | | - Béla Varga
- L2C, Univ Montpellier, CNRS, Montpellier, France
| | | | - Yannick Nicolas Gerber
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France; IUF, Intitut Universitaire de, France, Paris
| | | | | |
Collapse
|
4
|
Wang Z, Zhang Y, Wang L, Ito Y, Li G, Zhang P. Nerve implants with bioactive interfaces enhance neurite outgrowth and nerve regeneration in vivo. Colloids Surf B Biointerfaces 2022; 218:112731. [PMID: 35917689 DOI: 10.1016/j.colsurfb.2022.112731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/08/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
Abstract
Nerve implants functionalized with growth factors and stem cells are critical to promote neurite outgrowth, regulate neurodifferentiation, and facilitate nerve regeneration. In this study, human umbilical cord mesenchymal stem cells (hUCMSCs) and 3,4-hydroxyphenalyalanine (DOPA)-containing insulin-like growth factor 1 (DOPA-IGF-1) were simultaneously applied to enhance the bioactivity of poly(lactide-co-glycolide) (PLGA) substrates which will be potentially utilized as nerve implants. In vitro and in vivo evaluations indicated that hUCMSCs and DOPA-IGF-1 could synergistically regulate neurite outgrowth of PC12 cells, improve intravital recovery of motor functions, and promote conduction of nerve electrical signals in vivo. The enhanced functional and structural nerve regeneration of injured spinal cord might be mainly attributable to the synergistically enhanced biofunctionality of hUCMSCs and DOPA-IGF-1/PLGA on the bioactive interfaces. Findings from this study demonstrate the potential of hUCMSC-seeded, DOPA-IGF-1-modified PLGA implants as promising candidates for promoting axonal regeneration and motor functional recovery in spinal cord injury treatment.
Collapse
Affiliation(s)
- Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Yi Zhang
- Department of Urology, The Second Hospital, Jilin University, Changchun 130041, PR China
| | - Liqiang Wang
- Department of Ophthalmology, Third Medical Center, Chinese PLA General Hospital, Beijing 100853, PR China
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, Saitama 351-0198, Japan
| | - Gang Li
- Department of Orthopaedics and Traumatology and Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| |
Collapse
|
5
|
Coppola F, Russo T, Soares AMVM, Marques PAAP, Polese G, Pereira E, Freitas R. The influence of salinity on the toxicity of remediated seawater. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:32967-32987. [PMID: 35022978 DOI: 10.1007/s11356-021-17745-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/21/2021] [Indexed: 06/14/2023]
Abstract
Mercury (Hg) is one of the most hazardous pollutants, due to its toxicity, biological magnification and worldwide persistence in aquatic systems. Thus, new efficient nanotechnologies (e.g. graphene oxide functionalized with polyethyleneimine (GO-PEI)) have been developed to remove this metal from the water. Aquatic environments, in particular transitional systems, are also subjected to disturbances resulting from climate change, such as salinity shifts. Salinity is one of the most relevant factors that influences the distribution and survival of aquatic species such as mussels. To our knowledge, no studies assessed the ecotoxicological impairments induced in marine organisms exposed to remediate seawater (RSW) under different salinity levels. For this, the focus of the present study was to evaluate the effects of seawater previously contaminated with Hg and remediated with GO-PEI, using the species Mytilus galloprovincialis, maintained at three different salinities (30, 20 and 40). The results obtained demonstrated similar histopathological and metabolic alterations, oxidative stress and neurotoxicity in mussels under RSW treatment at stressful salinity conditions (20 and 40) in comparison to control salinity (30). On the other hand, the present findings revealed toxicological effects including cellular damage and histopathological impairments in mussels exposed to Hg contaminated seawater in comparison to non-contaminated ones, at each salinity level. Overall, these results confirm the high efficiency of GO-PEI to sorb Hg from water with no noticeable toxic effects even under different salinities, leading to consider it a promising eco-friendly approach to remediate contaminated water.
Collapse
Affiliation(s)
- Francesca Coppola
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Tania Russo
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Amadeu M V M Soares
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Paula A A P Marques
- Department of Mechanical Engineering & TEMA, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Gianluca Polese
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Eduarda Pereira
- Department of Chemistry & LAQV-REQUIMTE, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rosa Freitas
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
6
|
Zawadzka M, Kwaśniewska A, Miazga K, Sławińska U. Perspectives in the Cell-Based Therapies of Various Aspects of the Spinal Cord Injury-Associated Pathologies: Lessons from the Animal Models. Cells 2021; 10:cells10112995. [PMID: 34831217 PMCID: PMC8616284 DOI: 10.3390/cells10112995] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic injury of the spinal cord (SCI) is a devastating neurological condition often leading to severe dysfunctions, therefore an improvement in clinical treatment for SCI patients is urgently needed. The potential benefits of transplantation of various cell types into the injured spinal cord have been intensively investigated in preclinical SCI models and clinical trials. Despite the many challenges that are still ahead, cell transplantation alone or in combination with other factors, such as artificial matrices, seems to be the most promising perspective. Here, we reviewed recent advances in cell-based experimental strategies supporting or restoring the function of the injured spinal cord with a particular focus on the regenerative mechanisms that could define their clinical translation.
Collapse
|
7
|
Roy A, Pathak Z, Kumar H. Strategies to neutralize RhoA/ROCK pathway after spinal cord injury. Exp Neurol 2021; 343:113794. [PMID: 34166685 DOI: 10.1016/j.expneurol.2021.113794] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/01/2021] [Accepted: 06/19/2021] [Indexed: 01/22/2023]
Abstract
Regeneration is bungled following CNS injuries, including spinal cord injury (SCI). Inherent decay of permissive conditions restricts the regrowth of the mature CNS after an injury. Hypertrophic scarring, insignificant intrinsic axon-growth activity, and axon-growth inhibitory molecules such as myelin inhibitors and scar inhibitors constitute a significant hindrance to spinal cord repair. Besides these molecules, a combined absence of various mechanisms responsible for axonal regeneration is the main reason behind the dereliction of the adult CNS to regenerate. The neutralization of specific inhibitors/proteins by stymieing antibodies or encouraging enzymatic degradation results in improved axon regeneration. Previous efforts to induce regeneration after SCI have stimulated axonal development in or near lesion sites, but not beyond them. Several pathways are responsible for the axonal growth obstruction after a CNS injury, including SCI. Herein, we summarize the axonal, glial, and intrinsic factor which impedes the regeneration. We have also discussed the methods to stabilize microtubules and through this to maintain the proper cytoskeletal dynamics of growth cone as disorganized microtubules lead to the failure of axonal regeneration. Moreover, we primarily focus on diverse inhibitors of axonal growth and molecular approaches to counteract them and their downstream intracellular signaling through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Zarna Pathak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
8
|
Ribas VT, Vahsen BF, Tatenhorst L, Estrada V, Dambeck V, Almeida RA, Bähr M, Michel U, Koch JC, Müller HW, Lingor P. AAV-mediated inhibition of ULK1 promotes axonal regeneration in the central nervous system in vitro and in vivo. Cell Death Dis 2021; 12:213. [PMID: 33637688 PMCID: PMC7910615 DOI: 10.1038/s41419-021-03503-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 01/31/2023]
Abstract
Axonal damage is an early step in traumatic and neurodegenerative disorders of the central nervous system (CNS). Damaged axons are not able to regenerate sufficiently in the adult mammalian CNS, leading to permanent neurological deficits. Recently, we showed that inhibition of the autophagic protein ULK1 promotes neuroprotection in different models of neurodegeneration. Moreover, we demonstrated previously that axonal protection improves regeneration of lesioned axons. However, whether axonal protection mediated by ULK1 inhibition could also improve axonal regeneration is unknown. Here, we used an adeno-associated viral (AAV) vector to express a dominant-negative form of ULK1 (AAV.ULK1.DN) and investigated its effects on axonal regeneration in the CNS. We show that AAV.ULK1.DN fosters axonal regeneration and enhances neurite outgrowth in vitro. In addition, AAV.ULK1.DN increases neuronal survival and enhances axonal regeneration after optic nerve lesion, and promotes long-term axonal protection after spinal cord injury (SCI) in vivo. Interestingly, AAV.ULK1.DN also increases serotonergic and dopaminergic axon sprouting after SCI. Mechanistically, AAV.ULK1.DN leads to increased ERK1 activation and reduced expression of RhoA and ROCK2. Our findings outline ULK1 as a key regulator of axonal degeneration and regeneration, and define ULK1 as a promising target to promote neuroprotection and regeneration in the CNS.
Collapse
Affiliation(s)
- Vinicius Toledo Ribas
- Department of Morphology, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, 31270-901, Brazil.
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.
| | - Björn Friedhelm Vahsen
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Lars Tatenhorst
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Von-Siebold-Straße 3a, 37075, Göttingen, Germany
- DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Veronica Estrada
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Vivian Dambeck
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Von-Siebold-Straße 3a, 37075, Göttingen, Germany
- DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Raquel Alves Almeida
- Department of Morphology, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, 31270-901, Brazil
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Uwe Michel
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Jan Christoph Koch
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Hans Werner Müller
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Paul Lingor
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Von-Siebold-Straße 3a, 37075, Göttingen, Germany
- DFG Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- Department of Neurology, Rechts der Isar Hospital of the Technical University Munich, Ismaninger Straße 22, 81675, Munich, Germany
| |
Collapse
|
9
|
Heindryckx F, Li JP. Role of proteoglycans in neuro-inflammation and central nervous system fibrosis. Matrix Biol 2018; 68-69:589-601. [PMID: 29382609 DOI: 10.1016/j.matbio.2018.01.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/26/2017] [Accepted: 01/20/2018] [Indexed: 12/19/2022]
Abstract
Fibrosis is defined as the thickening and scarring of connective tissue, usually as a consequence of tissue damage. The central nervous system (CNS) is special in the sense that fibrogenic cells are restricted to vascular and meningeal areas. Inflammation and the disruption of the blood-brain barrier can lead to the infiltration of fibroblasts and trigger fibrotic response. While the initial function of the fibrotic tissue is to restore the blood-brain barrier and to limit the site of injury, it also demolishes the structure of extracellular matrix and impedes the healing process by producing inhibitory molecules and forming a physical and biochemical barrier that prevents axon regeneration. As a major constituent in the extracellular matrix, proteoglycans participate in the neuro-inflammation, modulating the fibrotic process. In this review, we will discuss the pathophysiology of fibrosis during acute injuries of the CNS, as well as during chronic neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease, multiple sclerosis and age-related neurodegeneration with focus on the functional roles of proteoglycans.
Collapse
Affiliation(s)
- Femke Heindryckx
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology/SciLifeLab, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
10
|
Kjell J, Olson L. Rat models of spinal cord injury: from pathology to potential therapies. Dis Model Mech 2017; 9:1125-1137. [PMID: 27736748 PMCID: PMC5087825 DOI: 10.1242/dmm.025833] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A long-standing goal of spinal cord injury research is to develop effective spinal cord repair strategies for the clinic. Rat models of spinal cord injury provide an important mammalian model in which to evaluate treatment strategies and to understand the pathological basis of spinal cord injuries. These models have facilitated the development of robust tests for assessing the recovery of locomotor and sensory functions. Rat models have also allowed us to understand how neuronal circuitry changes following spinal cord injury and how recovery could be promoted by enhancing spontaneous regenerative mechanisms and by counteracting intrinsic inhibitory factors. Rat studies have also revealed possible routes to rescuing circuitry and cells in the acute stage of injury. Spatiotemporal and functional studies in these models highlight the therapeutic potential of manipulating inflammation, scarring and myelination. In addition, potential replacement therapies for spinal cord injury, including grafts and bridges, stem primarily from rat studies. Here, we discuss advantages and disadvantages of rat experimental spinal cord injury models and summarize knowledge gained from these models. We also discuss how an emerging understanding of different forms of injury, their pathology and degree of recovery has inspired numerous treatment strategies, some of which have led to clinical trials. Summary: In this Review, we discuss the advantages and disadvantages of the rat for studies of experimental spinal cord injury and summarize the knowledge gained from such studies.
Collapse
Affiliation(s)
- Jacob Kjell
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich 80336, Germany
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
11
|
Ahn M, Moon C, Park C, Kim J, Sim KB, Shin T. Transient activation of an adaptor protein, disabled-2, in rat spinal cord injury. Acta Histochem 2015; 117:56-61. [PMID: 25432322 DOI: 10.1016/j.acthis.2014.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/04/2014] [Indexed: 12/01/2022]
Abstract
We previously reported that disabled-2 (Dab-2), a cytosolic adaptor protein, was expressed in inflammatory and glial cells in the central nervous system (CNS) in experimental autoimmune encephalomyelitis and cerebral cryoinjury. Here, to determine the pattern of Dab-2 expression in a clip compression-induced rat spinal cord injury (SCI) model, the protein level and localization of Dab-2 in the spinal cord were investigated in rats with SCI using Western blotting and immunohistochemistry. Western blotting revealed that the expression of both the 75- and 100-kDa isoforms of Dab-2 peaked significantly in the spinal cord after clip compression injury 7 days post-injury compared to sham controls, and declined slightly thereafter. Immunohistochemistry revealed weak Dab-2 immunostaining in some neurons, glial cells, and ependymal cells in the spinal cords of the control animals, compared to staining in the macrophages and reactive astrocytes in lesions of the SCI animals. Overall, these findings suggest that both isoforms of Dab-2 are transiently upregulated in response to SCI and that the increased expression of Dab-2 is associated with the early activation of macrophages and astrogliosis in the course of CNS inflammation.
Collapse
|
12
|
Niapour N, Niapour A, Sheikhkanloui Milan H, Amani M, Salehi H, Najafzadeh N, Gholami MR. All trans retinoic acid modulates peripheral nerve fibroblasts viability and apoptosis. Tissue Cell 2014; 47:61-5. [PMID: 25532484 DOI: 10.1016/j.tice.2014.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 11/02/2014] [Accepted: 11/17/2014] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Following peripheral nerve injury, residing fibroblasts start to proliferate and accumulate at the injury site and may participate in neuroma tissue evolution. Retinoic acid has been shown to regulate many cellular processes and to display anti-proliferative and anti-fibrotic properties. The aim of this study was to investigate the impact of all trans retinoic acid (ATRA) on rat peripheral nerve fibroblasts. MATERIALS AND METHODS Peripheral nerve fibroblasts and C166 cells were treated with increasing doses of ATRA (0.05 nM to 1 μM). The viability of cells was determined with 3-(4,5-dimethlthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. In addition, the number of peripheral nerve fibroblasts was counted after two days of ATRA treatment and alternatively up to the end of next week. Acridine orange/ethidium bromide double staining was implemented to morphologically visualize the possible mechanism of cell death. For apoptosis, caspase 3/7 activity was measured using Caspase-Glo 3/7 assay kit. RESULTS MTT assay revealed that 0.05-1 nM of ATRA reduces fibroblasts viabilities. Then, almost a plateau state was observed from 1 nM to 1 μM of ATRA exposure. Additionally, a deceleration in peripheral nerve fibroblasts growth was confirmed via cell counting. Quantification of acridine orange/ethidium bromide staining displayed highly increased number of early apoptotic cells following ATRA administration. Amplified activation of caspase 3/7 was in favor of apoptosis in ATRA treated peripheral nerve fibroblasts. CONCLUSION The data from the present study demonstrate that ATRA could interfere in peripheral nerve fibroblasts viabilities and induce apoptosis. Although more investigations are needed to be implemented, our in vitro results indicate that retinoic acid can probably help the regeneration of injured axon via reducing of fibroblasts growth.
Collapse
Affiliation(s)
- Nazila Niapour
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Niapour
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | | | - Mohammad Amani
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nowrouz Najafzadeh
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Gholami
- Department of Anatomical Sciences, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
13
|
Hwang DW, Jin Y, Lee DH, Kim HY, Cho HN, Chung HJ, Park Y, Youn H, Lee SJ, Lee HJ, Kim SU, Wang KC, Lee DS. In vivo bioluminescence imaging for prolonged survival of transplanted human neural stem cells using 3D biocompatible scaffold in corticectomized rat model. PLoS One 2014; 9:e105129. [PMID: 25198726 PMCID: PMC4157740 DOI: 10.1371/journal.pone.0105129] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 07/21/2014] [Indexed: 11/27/2022] Open
Abstract
Stem cell-based treatment of traumatic brain injury has been limited in its capacity to bring about complete functional recovery, because of the poor survival rate of the implanted stem cells. It is known that biocompatible biomaterials play a critical role in enhancing survival and proliferation of transplanted stem cells via provision of mechanical support. In this study, we noninvasively monitored in vivo behavior of implanted neural stem cells embedded within poly-l-lactic acid (PLLA) scaffold, and showed that they survived over prolonged periods in corticectomized rat model. Corticectomized rat models were established by motor-cortex ablation of the rat. F3 cells expressing enhanced firefly luciferase (F3-effLuc) were established through retroviral infection. The F3-effLuc within PLLA was monitored using IVIS-100 imaging system 7 days after corticectomized surgery. F3-effLuc within PLLA robustly adhered, and gradually increased luciferase signals of F3-effLuc within PLLA were detected in a day dependent manner. The implantation of F3-effLuc cells/PLLA complex into corticectomized rats showed longer-lasting luciferase activity than F3-effLuc cells alone. The bioluminescence signals from the PLLA-encapsulated cells were maintained for 14 days, compared with 8 days for the non-encapsulated cells. Immunostaining results revealed expression of the early neuronal marker, Tuj-1, in PLLA-F3-effLuc cells in the motor-cortex-ablated area. We observed noninvasively that the mechanical support by PLLA scaffold increased the survival of implanted neural stem cells in the corticectomized rat. The image-guided approach easily proved that scaffolds could provide supportive effect to implanted cells, increasing their viability in terms of enhancing therapeutic efficacy of stem-cell therapy.
Collapse
Affiliation(s)
- Do Won Hwang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Science, WCU Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Yeona Jin
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Do Hun Lee
- University of Miami School of Medicine, Miami Project to Cure Paralysis, Department of Neurological Surgery, Miami, Florida, United States of America
| | - Han Young Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Science, WCU Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Han Na Cho
- College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hye Jin Chung
- College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Yunwoong Park
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Imaging Center, Seoul National University Cancer Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Jin Lee
- College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hong J. Lee
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | - Seung U. Kim
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | - Kyu-Chang Wang
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Korea
- * E-mail: (DSL); (K-CW)
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Science, WCU Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
- * E-mail: (DSL); (K-CW)
| |
Collapse
|
14
|
Shrestha B, Coykendall K, Li Y, Moon A, Priyadarshani P, Yao L. Repair of injured spinal cord using biomaterial scaffolds and stem cells. Stem Cell Res Ther 2014; 5:91. [PMID: 25157690 PMCID: PMC4282172 DOI: 10.1186/scrt480] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The loss of neurons and degeneration of axons after spinal cord injury result in the loss of sensory and motor functions. A bridging biomaterial construct that allows the axons to grow through has been investigated for the repair of injured spinal cord. Due to the hostility of the microenvironment in the lesion, multiple conditions need to be fulfilled to achieve improved functional recovery. A scaffold has been applied to bridge the gap of the lesion as contact guidance for axonal growth and to act as a vehicle to deliver stem cells in order to modify the microenvironment. Stem cells may improve functional recovery of the injured spinal cord by providing trophic support or directly replacing neurons and their support cells. Neural stem cells and mesenchymal stem cells have been seeded into biomaterial scaffolds and investigated for spinal cord regeneration. Both natural and synthetic biomaterials have increased stem cell survival in vivo by providing the cells with a controlled microenvironment in which cell growth and differentiation are facilitated. This optimal multi‒disciplinary approach of combining biomaterials, stem cells, and biomolecules offers a promising treatment for the injured spinal cord.
Collapse
|
15
|
Scar-modulating treatments for central nervous system injury. Neurosci Bull 2014; 30:967-984. [PMID: 24957881 DOI: 10.1007/s12264-013-1456-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 04/09/2014] [Indexed: 02/04/2023] Open
Abstract
Traumatic injury to the adult mammalian central nervous system (CNS) leads to complex cellular responses. Among them, the scar tissue formed is generally recognized as a major obstacle to CNS repair, both by the production of inhibitory molecules and by the physical impedance of axon regrowth. Therefore, scar-modulating treatments have become a leading therapeutic intervention for CNS injury. To date, a variety of biological and pharmaceutical treatments, targeting scar modulation, have been tested in animal models of CNS injury, and a few are likely to enter clinical trials. In this review, we summarize current knowledge of the scar-modulating treatments according to their specific aims: (1) inhibition of glial and fibrotic scar formation, and (2) blockade of the production of scar-associated inhibitory molecules. The removal of existing scar tissue is also discussed as a treatment of choice. It is believed that only a combinatorial strategy is likely to help eliminate the detrimental effects of scar tissue on CNS repair.
Collapse
|
16
|
Estrada V, Brazda N, Schmitz C, Heller S, Blazyca H, Martini R, Müller HW. Long-lasting significant functional improvement in chronic severe spinal cord injury following scar resection and polyethylene glycol implantation. Neurobiol Dis 2014; 67:165-79. [PMID: 24713436 DOI: 10.1016/j.nbd.2014.03.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/17/2014] [Accepted: 03/28/2014] [Indexed: 01/04/2023] Open
Abstract
We identified a suitable biomatrix that improved axon regeneration and functional outcome after partial (moderate) and complete (severe) chronic spinal cord injury (SCI) in rat. Five weeks after dorsal thoracic hemisection injury the lesion scar was resected via aspiration and the resulting cavity was filled with different biopolymers such as Matrigel™, alginate-hydrogel and polyethylene glycol 600 (PEG) all of which have not previously been used as sole graft-materials in chronic SCI. Immunohistological staining revealed marked differences between these compounds regarding axon regeneration, invasion/elongation of astrocytes, fibroblasts, endothelial and Schwann cells, revascularization, and collagen deposition. According to axon regeneration-supporting effects, the biopolymers could be ranked in the order PEG>>alginate-hydrogel>Matrigel™. Even after complete chronic transection, the PEG-bridge allowed long-distance axon regeneration through the grafted area and for, at least, 1cm beyond the lesion/graft border. As revealed by electron microscopy, bundles of regenerating axons within the matrix area received myelin ensheathment from Schwann cells. The beneficial effects of PEG-implantation into the resection-cavity were accompanied by long-lasting significant locomotor improvement over a period of 8months. Following complete spinal re-transection at the rostral border of the PEG-graft the locomotor recovery was aborted, suggesting a functional role of regenerated axons in the initial locomotor improvement. In conclusion, scar resection and subsequent implantation of PEG into the generated cavity leads to tissue recovery, axon regeneration, myelination and functional improvement that have not been achieved before in severe chronic SCI.
Collapse
Affiliation(s)
- Veronica Estrada
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Nicole Brazda
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Christine Schmitz
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Silja Heller
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Heinrich Blazyca
- Department of Neurology, Developmental Neurobiology, University Medical Center Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Rudolf Martini
- Department of Neurology, Developmental Neurobiology, University Medical Center Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Hans Werner Müller
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
17
|
Burnside ER, Bradbury EJ. Review: Manipulating the extracellular matrix and its role in brain and spinal cord plasticity and repair. Neuropathol Appl Neurobiol 2014; 40:26-59. [DOI: 10.1111/nan.12114] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/20/2013] [Indexed: 12/17/2022]
Affiliation(s)
- E. R. Burnside
- King's College London; Regeneration Group; The Wolfson Centre for Age-Related Diseases; Guy's Campus; London UK
| | - E. J. Bradbury
- King's College London; Regeneration Group; The Wolfson Centre for Age-Related Diseases; Guy's Campus; London UK
| |
Collapse
|
18
|
Zhang YP, Walker MJ, Shields LBE, Wang X, Walker CL, Xu XM, Shields CB. Controlled cervical laceration injury in mice. J Vis Exp 2013:e50030. [PMID: 23685551 PMCID: PMC3679671 DOI: 10.3791/50030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Use of genetically modified mice enhances our understanding of molecular mechanisms underlying several neurological disorders such as a spinal cord injury (SCI). Freehand manual control used to produce a laceration model of SCI creates inconsistent injuries often associated with a crush or contusion component and, therefore, a novel technique was developed. Our model of cervical laceration SCI has resolved inherent difficulties with the freehand method by incorporating 1) cervical vertebral stabilization by vertebral facet fixation, 2) enhanced spinal cord exposure, and 3) creation of a reproducible laceration of the spinal cord using an oscillating blade with an accuracy of ±0.01 mm in depth without associated contusion. Compared to the standard methods of creating a SCI laceration such as freehand use of a scalpel or scissors, our method has produced a consistent lesion. This method is useful for studies on axonal regeneration of corticospinal, rubrospinal, and dorsal ascending tracts.
Collapse
Affiliation(s)
- Yi Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Liang H, Li C, Gao A, Liang P, Shao Y, Lin T, Zhang X. Spinal duraplasty with two novel substitutes restored locomotor function after acute laceration spinal cord injury in rats. J Biomed Mater Res B Appl Biomater 2012; 100:2131-40. [DOI: 10.1002/jbm.b.32778] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 05/29/2012] [Accepted: 06/28/2012] [Indexed: 02/07/2023]
|
20
|
Saxena T, Gilbert J, Stelzner D, Hasenwinkel J. Mechanical characterization of the injured spinal cord after lateral spinal hemisection injury in the rat. J Neurotrauma 2012; 29:1747-57. [PMID: 22435754 DOI: 10.1089/neu.2011.1818] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The glial scar formed at the site of traumatic spinal cord injury (SCI) has been classically hypothesized to be a potent physical and biochemical barrier to nerve regeneration. One longstanding hypothesis is that the scar acts as a physical barrier due to its increased stiffness in comparison to uninjured spinal cord tissue. However, the information regarding the mechanical properties of the glial scar in the current literature is mostly anecdotal and not well quantified. We monitored the mechanical relaxation behavior of injured rat spinal cord tissue at the site of mid-thoracic spinal hemisection 2 weeks and 8 weeks post-injury using a microindentation test method. Elastic moduli were calculated and a modified standard linear model (mSLM) was fit to the data to estimate the relaxation time constant and viscosity. The SLM was modified to account for a spectrum of relaxation times, a phenomenon common to biological tissues, by incorporating a stretched exponential term. Injured tissue exhibited significantly lower stiffness and elastic modulus in comparison to uninjured control tissue, and the results from the model parameters indicated that the relaxation time constant and viscosity of injured tissue were significantly higher than controls. This study presents direct micromechanical measurements of injured spinal cord tissue post-injury. The results of this study show that the injured spinal tissue displays complex viscoelastic behavior, likely indicating changes in tissue permeability and diffusivity.
Collapse
Affiliation(s)
- Tarun Saxena
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, USA
| | | | | | | |
Collapse
|
21
|
Park SS, Lee YJ, Lee SH, Lee D, Choi K, Kim WH, Kweon OK, Han HJ. Functional recovery after spinal cord injury in dogs treated with a combination of Matrigel and neural-induced adipose-derived mesenchymal Stem cells. Cytotherapy 2012; 14:584-97. [PMID: 22348702 DOI: 10.3109/14653249.2012.658913] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Previous studies have reported that scaffold or cell-based transplantation may improve functional recovery following spinal cord injury (SCI), but these results were based on neuronal regeneration and cell replacement. In this study, we investigated whether a combination of Matrigel and neural-induced mesenchymal stem cells (NMSC) improved hindlimb function in dogs with SCI, and what mechanisms were involved. METHODS We pre-differentiated canine adipose-derived mesenchymal stem cells into NMSC. A total of 12 dogs subjected to SCI procedures were assigned to one of the following three transplantation treatment groups: phosphate-buffered saline (PBS); Matrigel; or Matrigel seeded with NMSC. Treatment occurred 1 week after SCI. Basso, Beattie and Bresnahan (B.B.B.) and Tarlov scores, histopathology, immunofluorescence staining and Western blot analysis were used to evaluate the treatment effects. RESULTS Compared with dogs administered PBS or Matrigel alone, dogs treated with Matrigel + NMSC showed significantly better functional recovery 8 weeks after transplantation. Histology and immunochemical analysis revealed that the combination of Matrigel + NMSC reduced fibrosis from secondary injury processes and improved neuronal regeneration more than the other treatments. In addition, the combination of Matrigel + NMSC decreased the expression of inflammation and/or astrogliosis markers. Increased expressions of intracellular molecules related to neuronal extension, neuronal markers and neurotrophic factors were also found in the Matrigel + NMSC group. However, the expression of nestin as a neural stem cell marker was increased with Matrigel alone. CONCLUSIONS The combination of Matrigel + NMSC produced beneficial effects in dogs with regard to functional recovery following SCI through enhancement of anti-inflammation, anti-astrogliosis, neuronal extension and neuronal regeneration effects.
Collapse
Affiliation(s)
- Sung-Su Park
- Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Fawcett JW, Schwab ME, Montani L, Brazda N, Müller HW. Defeating inhibition of regeneration by scar and myelin components. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:503-22. [PMID: 23098733 DOI: 10.1016/b978-0-444-52137-8.00031-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Axon regeneration and the sprouting processes that underlie plasticity are blocked by inhibitory factors in the central nervous system (CNS) environment, several of which are upregulated after injury. The major inhibitory molecules are those associated with myelin and those associated with the glial scar. In myelin, NogoA, MAG, and OMgp are present on normal oligodendrocytes and on myelin debris. They act partly via the Nogo receptor, partly via an unidentified amino-Nogo receptor. In the glial scar, chondroitin sulphate proteoglycans, semaphorins, and the formation of a collagen-based membrane are all inhibitory. Methods to counteract these forms of inhibition have been identified, and these treatments promote axon regeneration in the damaged spinal cord, and in some cases recovery of function through enhanced plasticity.
Collapse
Affiliation(s)
- James W Fawcett
- Cambridge University Centre for Brain Repair, Cambridge, UK.
| | | | | | | | | |
Collapse
|
23
|
Agmatine-reduced collagen scar area accompanied with surface righting reflex recovery after complete transection spinal cord injury. Spine (Phila Pa 1976) 2011; 36:2130-8. [PMID: 21325984 DOI: 10.1097/brs.0b013e318205e3f7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Intended to investigate whether agmatine treatment reduces collagen scar area in mice subjected to spinal cord injury (SCI). OBJECTIVE The purpose of the present study is to demonstrate the protective effect of agmatine on complete transection SCI mice. SUMMARY OF BACK GROUND DATA: The deposition of collagen that occurs at the lesion site, during the SCI, was well known. Agmatine has been reported to exert neuroprotective effect in various stress models including central nervous system injuries. In the present investigation, we hypothesized that agmatine treatment could rescue the mice subjected to SCI. METHODS Complete SCI was made at the T9 level. Agmatine was dissolved in normal saline (100 mg/kg, Sigma, St. Louis, MO) and given intraperitoneally 5 minutes after complete transection daily for 4 weeks (agmatine-treated mice, n = 30). Controls received normal saline in the same manner (experimental control, n = 30). Surface righting reflex test, expression of bone morphogenetic protein-7 (BMP-7), TGFβ-2 (transforming growth factor β-2), and collagen scar area were measured and the results were compared with Mann-Whitney U test using SAS. RESULTS Agmatine treatment improved the surface righting reflex of mice at 4 weeks after SCI (P = 0.030). The collagen scar, physical barrier to axon regeneration, was noticeably diminished by agmatine treatment at 4 weeks after SCI (P = 0.001). The expression of BMP-7, which is considered both neuroprotective and neuroregenerative, was increased in agmatine treatment group compared with experimental control group in the early stages after SCI (P = 0.015 at 1 day after SCI; P = 0.010 at 3 days; P = 0.035 at 1 week; P = 0.826 at 2 weeks). The expression of TGFβ-2 correlated with the deposition of the collagen matrix at the lesion site was decreased with agmatine treatment at 1 and 2 weeks after SCI (P = 0.001 at 1 week; P = 0.002 at 2 weeks). Survival rate was found to be higher in agmatine treatment group than in the experimental control group for 4 weeks after SCI (P = 0.076). CONCLUSION These results suggest that agmatine treatment could support neuroregeneration by reducing the collagen scar area through decreasing the expression of TGFβ-2 and increasing the expression of BMP-7 after SCI. Especially, the improved surface righting reflex of agmatine-treated group proposes that agmatine treatment have the potency to facilitate functional recovery after SCI. However, the drug (agmatine) warrants further investigation in higher mammals.
Collapse
|
24
|
Haist V, Ulrich R, Kalkuhl A, Deschl U, Baumgärtner W. Distinct spatio-temporal extracellular matrix accumulation within demyelinated spinal cord lesions in Theiler's murine encephalomyelitis. Brain Pathol 2011; 22:188-204. [PMID: 21767322 DOI: 10.1111/j.1750-3639.2011.00518.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The accumulation of extracellular matrix (ECM) and glial scar formation are considered important factors for the failure of regeneration in central nervous system (CNS) injury and multiple sclerosis. Theiler's murine encephalomyelitis (TME) as a model of multiple sclerosis served to evaluate the spatio-temporal course of ECM alterations in demyelinating conditions. Microarray analysis revealed only mildly upregulated gene expression of ECM molecules, their biosynthesis pathways and pro-fibrotic factors, while upregulation of matrix remodeling enzymes was more prominent. Immunohistochemistry demonstrated progressive accumulation of chondroitin sulfate proteoglycans, glycoproteins and collagens within demyelinated TME lesions, paralleling the development of astrogliosis. Deposition of collagen IV, laminin, perlecan and tenascin-C started 28 days postinfection (dpi), collagen I, decorin, entactin and neurocan accumulated from 56 dpi on, and fibronectin from 98 dpi on. The basement membrane (BM) molecules collagen IV, entactin, fibronectin, laminin and perlecan showed perivascular and parenchymal deposition, while the non-BM components collagen I, decorin, neurocan and tenascin-C only accumulated in a nonvascular pattern in demyelinated areas. Contrary, phosphacan expression progressively decreased during TME. The immunoreactivity of aggrecan and brevican remained unchanged. The spatio-temporal association of matrix accumulation with astrogliosis suggests a mainly astrocytic origin of ECM deposits, which in turn may contribute to remyelination failure in TME.
Collapse
Affiliation(s)
- Verena Haist
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | | | | | | |
Collapse
|
25
|
Schira J, Gasis M, Estrada V, Hendricks M, Schmitz C, Trapp T, Kruse F, Kögler G, Wernet P, Hartung HP, Müller HW. Significant clinical, neuropathological and behavioural recovery from acute spinal cord trauma by transplantation of a well-defined somatic stem cell from human umbilical cord blood. ACTA ACUST UNITED AC 2011; 135:431-46. [PMID: 21903726 DOI: 10.1093/brain/awr222] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Stem cell therapy is a potential treatment for spinal cord injury and different stem cell types have been grafted into animal models and humans suffering from spinal trauma. Due to inconsistent results, it is still an important and clinically relevant question which stem cell type will prove to be therapeutically effective. Thus far, stem cells of human sources grafted into spinal cord mostly included barely defined heterogeneous mesenchymal stem cell populations derived from bone marrow or umbilical cord blood. Here, we have transplanted a well-defined unrestricted somatic stem cell isolated from human umbilical cord blood into an acute traumatic spinal cord injury of adult immune suppressed rat. Grafting of unrestricted somatic stem cells into the vicinity of a dorsal hemisection injury at thoracic level eight resulted in hepatocyte growth factor-directed migration and accumulation within the lesion area, reduction in lesion size and augmented tissue sparing, enhanced axon regrowth and significant functional locomotor improvement as revealed by three behavioural tasks (open field Basso-Beattie-Bresnahan locomotor score, horizontal ladder walking test and CatWalk gait analysis). To accomplish the beneficial effects, neither neural differentiation nor long-lasting persistence of the grafted human stem cells appears to be required. The secretion of neurite outgrowth-promoting factors in vitro further suggests a paracrine function of unrestricted somatic stem cells in spinal cord injury. Given the highly supportive functional characteristics in spinal cord injury, production in virtually unlimited quantities at GMP grade and lack of ethical concerns, unrestricted somatic stem cells appear to be a highly suitable human stem cell source for clinical application in central nervous system injuries.
Collapse
Affiliation(s)
- Jessica Schira
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Centre Düsseldorf, Moorenstr. 5, 40223 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zukor KA, Kent DT, Odelberg SJ. Meningeal cells and glia establish a permissive environment for axon regeneration after spinal cord injury in newts. Neural Dev 2011; 6:1. [PMID: 21205291 PMCID: PMC3025934 DOI: 10.1186/1749-8104-6-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 01/04/2011] [Indexed: 11/10/2022] Open
Abstract
Background Newts have the remarkable ability to regenerate their spinal cords as adults. Their spinal cords regenerate with the regenerating tail after tail amputation, as well as after a gap-inducing spinal cord injury (SCI), such as a complete transection. While most studies on newt spinal cord regeneration have focused on events occurring after tail amputation, less attention has been given to events occurring after an SCI, a context that is more relevant to human SCI. Our goal was to use modern labeling and imaging techniques to observe axons regenerating across a complete transection injury and determine how cells and the extracellular matrix in the injury site might contribute to the regenerative process. Results We identify stages of axon regeneration following a spinal cord transection and find that axon regrowth across the lesion appears to be enabled, in part, because meningeal cells and glia form a permissive environment for axon regeneration. Meningeal and endothelial cells regenerate into the lesion first and are associated with a loose extracellular matrix that allows axon growth cone migration. This matrix, paradoxically, consists of both permissive and inhibitory proteins. Axons grow into the injury site next and are closely associated with meningeal cells and glial processes extending from cell bodies surrounding the central canal. Later, ependymal tubes lined with glia extend into the lesion as well. Finally, the meningeal cells, axons, and glia move as a unit to close the gap in the spinal cord. After crossing the injury site, axons travel through white matter to reach synaptic targets, and though ascending axons regenerate, sensory axons do not appear to be among them. This entire regenerative process occurs even in the presence of an inflammatory response. Conclusions These data reveal, in detail, the cellular and extracellular events that occur during newt spinal cord regeneration after a transection injury and uncover an important role for meningeal and glial cells in facilitating axon regeneration. Given that these cell types interact to form inhibitory barriers in mammals, identifying the mechanisms underlying their permissive behaviors in the newt will provide new insights for improving spinal cord regeneration in mammals.
Collapse
Affiliation(s)
- Katherine A Zukor
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT 84132, USA
| | | | | |
Collapse
|
27
|
Xu X, Geremia N, Bao F, Pniak A, Rossoni M, Brown A. Schwann cell coculture improves the therapeutic effect of bone marrow stromal cells on recovery in spinal cord-injured mice. Cell Transplant 2010; 20:1065-86. [PMID: 21092402 DOI: 10.3727/096368910x544906] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Studies of bone marrow stromal cells (MSCs) transplanted into the spinal cord-injured rat give mixed results: some groups report improved locomotor recovery while others only demonstrate improved histological appearance of the lesion. These studies show no clear correlation between neurological improvements and MSC survival. We examined whether MSC survival in the injured spinal cord could be enhanced by closely matching donor and recipient mice for genetic background and marker gene expression and whether exposure of MSCs to a neural environment (Schwann cells) prior to transplantation would improve their survival or therapeutic effects. Mice underwent a clip compression spinal cord injury at the fourth thoracic level and cell transplantation 7 days later. Despite genetic matching of donors and recipients, MSC survival in the injured spinal cord was very poor (∼1%). However, we noted improved locomotor recovery accompanied by improved histopathological appearance of the lesion in mice receiving MSC grafts. These mice had more white and gray matter sparing, laminin expression, Schwann cell infiltration, and preservation of neurofilament and 5-HT-positive fibers at and below the lesion. There was also decreased collagen and chondroitin sulphate proteoglycan deposition in the scar and macrophage activation in mice that received the MSC grafts. The Schwann cell cocultured MSCs had greater effects than untreated MSCs on all these indices of recovery. Analyses of chemokine and cytokine expression revealed that MSC/Schwann cell cocultures produced far less MCP-1 and IL-6 than MSCs or Schwann cells cultured alone. Thus, transplanted MSCs may improve recovery in spinal cord-injured mice through immunosuppressive effects that can be enhanced by a Schwann cell coculturing step. These results indicate that the temporary presence of MSCs in the injured cord is sufficient to alter the cascade of pathological events that normally occurs after spinal cord injury, generating a microenvironment that favors improved recovery.
Collapse
Affiliation(s)
- Xiaoyun Xu
- The Spinal Cord Injury Team, BioTherapeutics Research Laboratories and Molecular Brain Research Group, Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
28
|
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
|
29
|
Schiwy N, Brazda N, Müller HW. Enhanced regenerative axon growth of multiple fibre populations in traumatic spinal cord injury following scar-suppressing treatment. Eur J Neurosci 2009; 30:1544-53. [PMID: 19817844 DOI: 10.1111/j.1460-9568.2009.06929.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We analysed the effect of scar-suppressing treatment (anti-scarring treatment; AST) on augmenting axonal regeneration of various neuronal populations following spinal cord injury (SCI) in adult rat. AST included local iron chelator (2,2'-dipyridine-5,5'-dicarboxylic acid) injection and 8-bromo-cyclic adenosine monophosphate application to the lesion core. In previous studies, this treatment promoted long-distance regeneration of cut corticospinal tract axons, neuroprotection of projecting cortical neurons and functional improvement of treated rats [N. Klapka et al. (2005)Eur. J. Neurosci., 22, 3047-3058]. Treatment yielded significantly enhanced regrowth of descending serotonergic (5-HT), catecholaminergic (tyrosine hydroxylase; TH), corticospinal and rubrospinal axons into the lesion zone, as assessed by anterograde tracing and immunohistochemistry followed by quantification of axon profiles at 5 and 12 weeks post-injury. In addition, the determination of axons crossing the proximal borderline from uninjured tissue into fibrous scar area revealed a significant AST-promoted increase of intersecting fibres for 5-HT, TH and calcitonin gene-related peptide containing ascending sensory fibres. For a prolonged time period after lesion, the delayed (secondary) scar developing in treated rats is significantly more permeable for all analysed axon tracts than the initial (primary) scar forming in injured control animals lacking treatment. Furthermore, enhanced outgrowth of descending axons from fibrous scar into distal healthy spinal tissue was achieved in treated animals, and is in line with previous functional studies [S. Hermanns et al. (2001) Restor. Neurol. Neurosci., 19,139-148; N. Klapka et al. (2005)Eur. J. Neurosci., 22, 3047-3058]. Our findings indicate that AST exerts a prolonged beneficial effect on fibrous scarring allowing enhanced axonal regrowth of different fibre tracts in SCI regardless of their distinct regenerative demands.
Collapse
Affiliation(s)
- Nora Schiwy
- Molecular Neurobiology Laboratory, Heinrich-Heine-University Düsseldorf, 40223 Düsseldorf, Germany
| | | | | |
Collapse
|
30
|
Fouad K, Ghosh M, Vavrek R, Tse AD, Pearse DD. Dose and chemical modification considerations for continuous cyclic AMP analog delivery to the injured CNS. J Neurotrauma 2009; 26:733-40. [PMID: 19397425 DOI: 10.1089/neu.2008.0730] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In this investigation, two cell-permeable synthetic analogs of cAMP, dibutyryl-cAMP (db-cAMP) and 8-bromo-cAMP, which are widely used to elevate intracellular cAMP levels under experimental conditions, were investigated for their ability to dose-dependently improve histological and functional outcomes following continuous delivery in two models of incomplete spinal cord injury (SCI). The cAMP analogs were delivered via osmotic minipumps at 1-250 mM through an indwelling cortical cannula or by intrathecal infusion for up to 4 weeks after either a T8 unilateral over-hemisection or a C2-3 dorsolateral quadrant lesion, respectively. In both SCI models, continuous db-cAMP delivery was associated with histopathological changes that included sporadic micro-hemorrhage formation and cavitation, enhanced macrophage infiltration and tissue damage at regions beyond the immediate application site; no deleterious or beneficial effect of agent delivery was observed at the spinal injury site. Furthermore, these changes were accompanied by pronounced behavioral deficits that included an absence of progressive locomotor recovery, increased extensor tone, paralysis, and sensory abnormalities. These deleterious effects were not observed in saline-treated animals, in animals in which the db-cAMP dose did not exceed 1 mM, or in those animals that received a high dose (250 mM) of the alternative cAMP analog, 8-bromo-cAMP. These results demonstrate that, for continuous intraparenchymal or intrathecal administration of cAMP analogs for the study of biological or therapeutic effects within the central nervous system (CNS), consideration of the effective concentration applied as well as the potential toxicity of chemical moieties on the parent molecule and/or their activity needs to be taken into account.
Collapse
Affiliation(s)
- Karim Fouad
- University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
31
|
Abstract
CNS injury-induced hemorrhage and tissue damage leads to excess iron, which can cause secondary degeneration. The mechanisms that handle this excess iron are not fully understood. We report that spinal cord contusion injury (SCI) in mice induces an "iron homeostatic response" that partially limits iron-catalyzed oxidative damage. We show that ceruloplasmin (Cp), a ferroxidase that oxidizes toxic ferrous iron, is important for this process. SCI in Cp-deficient mice demonstrates that Cp detoxifies and mobilizes iron and reduces secondary tissue degeneration and functional loss. Our results provide new insights into how astrocytes and macrophages handle iron after SCI. Importantly, we show that iron chelator treatment has a delayed effect in improving locomotor recovery between 3 and 6 weeks after SCI. These data reveal important aspects of the molecular control of CNS iron homeostasis after SCI and suggest that iron chelator therapy may improve functional recovery after CNS trauma and hemorrhagic stroke.
Collapse
|
32
|
Brazda N, Müller HW. Pharmacological modification of the extracellular matrix to promote regeneration of the injured brain and spinal cord. PROGRESS IN BRAIN RESEARCH 2009; 175:269-81. [PMID: 19660662 DOI: 10.1016/s0079-6123(09)17518-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
This chapter focuses on the role of the fibrous lesion scar as a major impediment for axonal regeneration in the injured central nervous system (CNS). We describe the appearance and complementary distribution of the glial and fibrous scar components in spinal cord lesions focusing on the morphology as well as on axon growth inhibitory molecular components accumulating in the collagenous and basement membrane-rich fibrous scar. We further report on the differential responses to fibrous scar of distinct fiber tracts in the injured spinal cord including the rubrospinal and corticospinal tracts as well as serotonergic, dopaminergic, and calcitonin gene-related peptide (CGRP) systems. Finally, we discuss therapeutic strategies to suppress fibrous scarring in traumatic CNS injury with particular emphasis on a unique pharmacological treatment using iron chelators and cyclic adenosine monophosphate (cAMP) to inhibit collagen biosynthesis. The latter treatment has been shown to promote long-distance axon growth, retrograde protection of injured neurons, and significant functional improvement.
Collapse
Affiliation(s)
- Nicole Brazda
- Department of Neurology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | | |
Collapse
|
33
|
Activation of the hypoxia-inducible factor-1alpha pathway accelerates bone regeneration. Proc Natl Acad Sci U S A 2008; 105:686-91. [PMID: 18184809 DOI: 10.1073/pnas.0708474105] [Citation(s) in RCA: 377] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The hypoxia-inducible factor-1alpha (HIF-1alpha) pathway is the central regulator of adaptive responses to low oxygen availability and is required for normal skeletal development. Here, we demonstrate that the HIF-1alpha pathway is activated during bone repair and can be manipulated genetically and pharmacologically to improve skeletal healing. Mice lacking pVHL in osteoblasts with constitutive HIF-1alpha activation in osteoblasts had markedly increased vascularity and produced more bone in response to distraction osteogenesis, whereas mice lacking HIF-1alpha in osteoblasts had impaired angiogenesis and bone healing. The increased vascularity and bone regeneration in the pVHL mutants were VEGF dependent and eliminated by concomitant administration of VEGF receptor antibodies. Small-molecule inhibitors of HIF prolyl hydroxylation stabilized HIF/VEGF production and increased angiogenesis in vitro. One of these molecules (DFO) administered in vivo into the distraction gap increased angiogenesis and markedly improved bone regeneration. These results identify the HIF-1alpha pathway as a critical mediator of neoangiogenesis required for skeletal regeneration and suggest the application of HIF activators as therapies to improve bone healing.
Collapse
|
34
|
Reynolds LF, Bren MC, Wilson BC, Gibson GD, Shoichet MS, Murphy RJL. Transplantation of porous tubes following spinal cord transection improves hindlimb function in the rat. Spinal Cord 2007; 46:58-64. [PMID: 17420773 DOI: 10.1038/sj.sc.3102063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN Experimental. OBJECTIVE To determine the effects of a porous tube transplant in spinal cord transected rats. SETTING Acadia University, Wolfville, Nova Scotia, Canada. METHODS Female rats were randomly assigned to three experimental groups: control (Con, n=8), spinal cord transected (Tx, n=5) and spinal cord transected with transplant (TxTp, n=7). The rats in the TxTp and Tx groups received a complete spinal cord transection at the T10 level and the TxTp group immediately received a porous tube transplant. RESULTS Locomotor activity rated on the Basso, Beattie, Bresnahan scale improved significantly in the TxTp animals over the 4 weeks such that final scores were 21, 1.4 and 7.1 for the Con, Tx and TxTp groups, respectively. As expected, the muscle to body mass ratios of the hindlimb skeletal muscles of the Tx group were decreased (soleus 35%, plantaris 29% and gastrocnemius 29%) and this was also observed in the TxTp group (soleus 33%, plantaris 23% and gastrocnemius 30%). Cytochrome c oxidase (CYTOX) activity in the plantaris was decreased by Tx but maintained in the TxTp group (Con=82.2, Tx=44.8 and TxTp=72.8 U/min/g). CONCLUSION Four weeks after the spinal cord transection, plantaris CYTOX activity and locomotor function improved with porous tube implantation. SPONSORSHIP Natural Sciences and Engineering Research Council.
Collapse
Affiliation(s)
- L F Reynolds
- School of Recreation Management and Kinesiology, Acadia University, Canada
| | | | | | | | | | | |
Collapse
|
35
|
Zurn AD, Bandtlow CE. Regeneration failure in the CNs: cellular and molecular mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:54-76. [PMID: 16955704 DOI: 10.1007/0-387-30128-3_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Anne D Zurn
- Department of Experimental Surgery, Lausanne University Hospital, Faculty of Biology and Medicine, Switzerland
| | | |
Collapse
|
36
|
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
|
37
|
Hendriks WTJ, Eggers R, Verhaagen J, Boer GJ. Gene transfer to the spinal cord neural scar with lentiviral vectors: predominant transgene expression in astrocytes but not in meningeal cells. J Neurosci Res 2007; 85:3041-52. [PMID: 17671987 DOI: 10.1002/jnr.21432] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Viral vector-mediated overexpression of neurotrophins in cells constituting the neural scar may represent a powerful approach to rendering scar tissue of a central nervous system (CNS) lesion permissive for neuronal regrowth. In this study a lentiviral vector encoding green fluorescent protein (LV-GFP) was injected in and around the neural scar 2 weeks after a dorsal column lesion in the rat spinal cord in order to analyze transduction characteristics of the neural scar after 4, 7, and 14 days. GFP expression was found at all points after injection and increased from 4 to 7 days, with no apparent difference observed between 7 and 14 days. The core of the lesion was virtually devoid of GFP signal despite direct vector injections in this area. The colocalization of GFP with specific cell markers (GFAP, vimentin, Raldh2, NeuN, OX-42, ED-1, and NG-2) indicated that the predominant cells transduced in the rim of the lesion were astrocytes, with neurons, microglia, oligodendrocyte precursors, and macrophages transduced to a lesser extent. None of the Raldh2-positive meningeal cells, present in the core of the scar, expressed GFP. In vitro meningeal cells were readily transduced, indicating that in vivo the formation of an extracellular matrix might prevent LV particles from transducing cells in the core of the scar. Because astrocytes are important cellular constituents of the glial scar after CNS injury, transduction of astrocytes with LV vectors encoding neurotrophic factors like BDNF or NT-3 may be used to enhance regeneration of severed axonal tracts through or along boundaries of a CNS lesion.
Collapse
Affiliation(s)
- W T J Hendriks
- Laboratory for Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | | | | | | |
Collapse
|
38
|
Iannotti C, Zhang YP, Shields LBE, Han Y, Burke DA, Xu XM, Shields CB. Dural repair reduces connective tissue scar invasion and cystic cavity formation after acute spinal cord laceration injury in adult rats. J Neurotrauma 2006; 23:853-65. [PMID: 16774471 DOI: 10.1089/neu.2006.23.853] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
This study examined whether duraplasty after acute cervical laceration spinal cord injury (SCI) in a rat model could (1) improve cerebrospinal fluid (CSF) circulation adjacent to the injury; (2) minimize connective tissue scarring; and (3) reduce post-traumatic inflammation and cystic cavitation. Following a transverse dural/arachnoid incision and C5-6 dorsal spinal hemisection, a 5-mm2 cadaveric dura mater allograft was placed over the lesion and fixed with fibrin glue (n=12). Control animals received an identical dural/arachnoid incision and cervical dorsal hemisection without dural repair (n=12). At 1, 5, and 10 weeks post-injury, plain film myelograms were obtained to characterize CSF circulation, and stereological methods were used to compare the extent of tissue sparing between the two groups. Immunohistochemical studies were performed to assess the degree of inflammation (ED-1), connective tissue scarring (laminin and type IV collagen), and reactive astrogliosis (GFAP). Our results indicate that dural allograft can improve CSF flow adjacent to the site of injury, which may be due to reduced meningeal fibrosis/scarring at the lesion site. Stereological analysis demonstrated that duraplasty resulted in a significant reduction in lesion volume at each time-point (p<0.01) associated with a nearly complete attenuation of post-traumatic cystic cavitation (p<0.001). Immunofluorescence studies demonstrated that duraplasty reduced the infiltration of ED-1-positive macrophages/microglia into and surrounding the lesion site, which may be responsible for the marked reduction in secondary injury following duraplasty. We conclude that duraplasty following acute spinal cord laceration may (1) improve CSF flow by limiting meningeal fibrosis; (2) reduce connective tissue scar formation; and (3) attenuate macrophage accumulation and progressive secondary injury.
Collapse
Affiliation(s)
- Christopher Iannotti
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville School of Medicine Louisville, Kentucky 40292, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The fibrous scar that develops after central nervous system (CNS) injury is considered a major impediment for axonal regeneration. It consists of a dense collagen IV meshwork, which serves as a binding matrix for numerous other extracellular matrix components and inhibitory molecules like proteoglycans and semaphorins, but also growth-promoting factors. Inhibition of collagen matrix formation in brain and spinal cord lesions leads to axonal regeneration and functional recovery, although collagen IV per se is not inhibitory for axonal outgrowth. This review focuses on the molecular properties of the collagen IV matrix and its interactions with various molecules that are expressed after CNS lesion. Moreover, studies on collagen expression and matrix formation after injury of regenerating versus non-regenerating nervous systems are reviewed. Major differences in collagen deposition in the CNS and the peripheral nervous system (PNS) and differences in specific cell responses to extracellular matrix deposition in the lesion area are discussed. Therapeutic treatments aiming at suppression of fibrous scarring have been shown to promote axon regeneration in various lesion paradigms of the mammalian CNS.
Collapse
Affiliation(s)
- Nicole Klapka
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine University, Düsseldorf, Germany
| | | |
Collapse
|
40
|
Regeneration and Repair. Dev Neurobiol 2006. [DOI: 10.1007/0-387-28117-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
41
|
Klapka N, Hermanns S, Straten G, Masanneck C, Duis S, Hamers FPT, Müller D, Zuschratter W, Müller HW. Suppression of fibrous scarring in spinal cord injury of rat promotes long-distance regeneration of corticospinal tract axons, rescue of primary motoneurons in somatosensory cortex and significant functional recovery. Eur J Neurosci 2006; 22:3047-58. [PMID: 16367771 DOI: 10.1111/j.1460-9568.2005.04495.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Traumatic injury of the central nervous system results in formation of a collagenous basement membrane-rich fibrous scar in the lesion centre. Due to accumulation of numerous axon-growth inhibitory molecules the lesion scar is considered a major impediment for axon regeneration. Following transection of the dorsal corticospinal tract (CST) at thoracic level 8 in adult rats, transient suppression of collagenous scarring in the lesion zone by local application of a potent iron chelator and cyclic adenosine monophosphate resulted in the delay of fibrous scarring. Treated animals displayed long-distance growth of CST axons through the lesion area extending for up to 1.5-2 cm into the distal cord. In addition, the treatment showed a strong neuroprotective effect, rescuing cortical motoneurons projecting into the CST that normally die (30%) after thoracic axotomy. Further, anterogradely traced CST axons regenerated through both grey and white matter and developed terminal arborizations in grey matter regions. In contrast to controls, injured animals receiving treatment showed significant functional recovery in the open field, in the horizontal ladder and in CatWalk locomotor tasks. We conclude that the fibrous lesion scar plays a pivotal role as a growth barrier for regenerating axons in adult spinal cord and that a delay in fibrotic scarring by local inhibition of collagen biosynthesis and basement membrane deposition is a promising and unique therapeutic strategy for treating human spinal trauma.
Collapse
Affiliation(s)
- Nicole Klapka
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Schwab JM, Brechtel K, Mueller CA, Failli V, Kaps HP, Tuli SK, Schluesener HJ. Experimental strategies to promote spinal cord regeneration--an integrative perspective. Prog Neurobiol 2006; 78:91-116. [PMID: 16487649 DOI: 10.1016/j.pneurobio.2005.12.004] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2005] [Revised: 12/13/2005] [Accepted: 12/13/2005] [Indexed: 11/17/2022]
Abstract
Detailed pathophysiological findings of secondary damage phenomena after spinal cord injury (SCI) as well as the identification of inhibitory and neurotrophic proteins have yielded a plethora of experimental therapeutic approaches. Main targets are (i) to minimize secondary damage progression (neuroprotection), (ii) to foster axon conduction (neurorestoration) and (iii) to supply a permissive environment to promote axonal sprouting (neuroregenerative therapies). Pre-clinical studies have raised hope in functional recovery through the antagonism of growth inhibitors, application of growth factors, cell transplantation, and vaccination strategies. To date, even though based on successful pre-clinical animal studies, results of clinical trials are characterized by dampened effects attributable to difficulties in the study design (patient heterogeneity) and species differences. A combination of complementary therapeutic strategies might be considered pre-requisite for future synergistic approaches. Here, we line out pre-clinical interventions resulting in improved functional neurological outcome after spinal cord injury and track them on their intended way to bedside.
Collapse
Affiliation(s)
- Jan M Schwab
- Institute of Brain Research, Calwer Str. 3, University of Tuebingen, Medical School, Calwerstr. 3, 72076 Tuebingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
43
|
Deumens R, Koopmans GC, Joosten EAJ. Regeneration of descending axon tracts after spinal cord injury. Prog Neurobiol 2005; 77:57-89. [PMID: 16271433 DOI: 10.1016/j.pneurobio.2005.10.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2005] [Revised: 08/23/2005] [Accepted: 10/05/2005] [Indexed: 02/03/2023]
Abstract
Axons within the adult mammalian central nervous system do not regenerate spontaneously after injury. Upon injury, the balance between growth promoting and growth inhibitory factors in the central nervous system dramatically changes resulting in the absence of regeneration. Axonal responses to injury vary considerably. In central nervous system regeneration studies, the spinal cord has received a lot of attention because of its relatively easy accessibility and its clinical relevance. The present review discusses the axon-tract-specific requirements for regeneration in the rat. This knowledge is very important for the development and optimalization of therapies to repair the injured spinal cord.
Collapse
Affiliation(s)
- Ronald Deumens
- Department of Psychiatry and Neuropsychology, Division Neuroscience, European Graduate School of Neuroscience EURON, University of Maastricht, Maastricht, The Netherlands.
| | | | | |
Collapse
|
44
|
Polikov VS, Tresco PA, Reichert WM. Response of brain tissue to chronically implanted neural electrodes. J Neurosci Methods 2005; 148:1-18. [PMID: 16198003 DOI: 10.1016/j.jneumeth.2005.08.015] [Citation(s) in RCA: 1112] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Revised: 08/06/2005] [Accepted: 08/08/2005] [Indexed: 11/16/2022]
Abstract
Chronically implanted recording electrode arrays linked to prosthetics have the potential to make positive impacts on patients suffering from full or partial paralysis. Such arrays are implanted into the patient's cortical tissue and record extracellular potentials from nearby neurons, allowing the information encoded by the neuronal discharges to control external devices. While such systems perform well during acute recordings, they often fail to function reliably in clinically relevant chronic settings. Available evidence suggests that a major failure mode of electrode arrays is the brain tissue reaction against these implants, making the biocompatibility of implanted electrodes a primary concern in device design. This review presents the biological components and time course of the acute and chronic tissue reaction in brain tissue, analyses the brain tissue response of current electrode systems, and comments on the various material science and bioactive strategies undertaken by electrode designers to enhance electrode performance.
Collapse
Affiliation(s)
- Vadim S Polikov
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | | |
Collapse
|
45
|
Walsh JF, Manwaring ME, Tresco PA. Directional Neurite Outgrowth Is Enhanced by Engineered Meningeal Cell-Coated Substrates. ACTA ACUST UNITED AC 2005; 11:1085-94. [PMID: 16144444 DOI: 10.1089/ten.2005.11.1085] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
After injury to the CNS, the anatomical organization of the tissue is disrupted, posing a barrier to the regeneration of axons. Meningeal cells, a central participant in the CNS tissue response to injury, migrate into the core of the wound site in an unorganized fashion and deposit a disorganized extracellular matrix (ECM) that produces a nonpermissive environment. Previous work in our laboratory has shown that the presentation of nanometer-scale topographic cues to these cells influences their morphological, cytoskeletal, and secreted ECM alignment. In the present study, we provided similar environmental cues to meningeal cells and examined the ability of the composite construct to influence dorsal root ganglion regeneration in vitro. When grown on control surfaces of meningeal cells lacking underlying topographic cues, there was no bias in neurite outgrowth. In contrast, when grown on monolayers of meningeal cells with underlying nanometer-scale topography, neurite outgrowth length was greater and was directed parallel to the underlying surface topography even though there exists an intervening meningeal cell layer. The observed outgrowth was significantly longer than on laminin-coated surfaces, which are considered to be the optimal substrata for promoting outgrowth of dorsal root ganglion neurons in culture. These results suggest that the nanometer-level surface finish of an implanted biomaterial may be used to organize the encapsulation tissue that accompanies the implantation of materials into the CNS. It furthermore suggests a simple approach for improving bridging materials for repair of nerve tracts or for affecting cellular organization at a device-tissue interface.
Collapse
Affiliation(s)
- Jennifer F Walsh
- Keck Center for Tissue Engineering, Department of Bioengineering, University of Utah, Salt Lake City, 84112, USA
| | | | | |
Collapse
|
46
|
Zhang YP, Iannotti C, Shields LBE, Han Y, Burke DA, Xu XM, Shields CB. Dural closure, cord approximation, and clot removal: enhancement of tissue sparing in a novel laceration spinal cord injury model. J Neurosurg 2004; 100:343-52. [PMID: 15070142 DOI: 10.3171/spi.2004.100.4.0343] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Laceration-induced spinal cord injury (SCI) results in the invasion of a connective tissue scar, progressive damage to the spinal cord due to complex secondary injury mechanisms, and axonal dieback of descending motor pathways. The authors propose that preparation of the spinal cord for repair strategies should include hematoma removal and dural closure, resulting in apposition of the severed ends of the spinal cord. Such procedures may reduce the size of the postinjury spinal cord cyst as well as limit scar formation. METHODS Using a novel device, the Vibraknife, the authors created a dorsal hemisection of the spinal cord at C-6 in the adult rat. In Group 1 (eight rats), the dura mater was repaired with apposition of the two stumps of the spinal cord to reduce the lesion gap. In Group 2 (10 rats), the dura was not closed and the two cord stumps were not approximated. All rats were killed at 4 weeks postinjury, and the spinal cords from each group were removed and examined using histological, stereological, and immunohistochemical methods. In Group 1 rats a significant reduction of the total lesion volume and connective tissue scar was observed compared with those in Group 2 (Student t-test, p < 0.05). Approximation of the stumps did not promote the regeneration of corticospinal tract fibers or sensory axons through the lesion site. CONCLUSIONS Apposition of the severed ends of the spinal cord by dural closure reduces the lesion gap, cystic cavitation, and connective tissue scar formation. These outcomes may collectively reduce secondary tissue damage at the injury site and shorten the length of the lesion gap, which will facilitate transplantation-mediated axonal regeneration after laceration-induced SCI.
Collapse
Affiliation(s)
- Yi Ping Zhang
- Department of Neurological Surgery, Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Abstract
Axon growth inhibitors associated with myelin and the glial scar contribute to the failure of axon regeneration in the injured adult mammalian central nervous system (CNS). A number of these inhibitors, their receptors, and signaling pathways have been identified. These inhibitors can now be neutralized by a variety of approaches that point to the possibility of developing new therapeutic strategies to stimulate regeneration after spinal cord injury.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, McGill University Health Centre, Montreal General Hospital Research Institute, 1650 Cedar Avenue, Montreal, Quebec, Canada, H3G 1A4.
| | | |
Collapse
|
49
|
Heuer H, Christ S, Friedrichsen S, Brauer D, Winckler M, Bauer K, Raivich G. Connective tissue growth factor: a novel marker of layer VII neurons in the rat cerebral cortex. Neuroscience 2003; 119:43-52. [PMID: 12763067 DOI: 10.1016/s0306-4522(03)00100-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Connective tissue growth factor (CTGF) belongs to a family of secreted, extracellular matrix-associated proteins that are involved in the regulation of cellular functions such as adhesion, migration, mitogenesis, differentiation and survival. Recent studies have also suggested the up-regulation of CTGF in response to trauma, scar formation and excitotoxicity in the CNS. To further elucidate the localization and regulation of this molecule in the rat brain we performed in situ hybridization experiments and found a very strong and selective expression of CTGF messenger ribonucleic acid (mRNA) on the band of layer VII neurons throughout the adult cerebral cortex. Similarly strong neuronal expression was also present in the dorsal endopiriform nucleus, extending rostrally from the ventrocaudal cortical layer VII, and in the deep layers of the olfactory glomeruli and the accessory olfactory nucleus. Double in situ hybridization confirmed selective CTGF mRNA expression on a subpopulation (approximately 35%) of microtubule-associated protein 2 mRNA-positive neurons in the cortical layer VII and the dorsal endopiriform nucleus. The nucleus of lateral olfactory tract showed moderate signal intensity; other parts of the forebrain, mesencephalon and brain stem only revealed a very weak level of CTGF mRNA expression. Non-neuronal expression was rare, considerably weaker than on cortical layer VII neurons, and normally associated with blood vessels. Developmental analysis of CTGF mRNA expression in embryonic and postnatal mouse also showed a moderately late onset at embryonic day 16-18, and confirmed the presence of CTGF mRNA in cortical layer VII in a second rodent species. Interestingly, injury experiments using direct cerebral trauma or injection of excitotoxic kainic acid into rat brain failed to up-regulate CTGF mRNA after injury and during the ensuing period of neuronal cell death, gliosis and neural scar tissue formation. Altogether, the current data suggest a constitutive role of CTGF, particularly in the adult cerebral cortex. In view of the strong ascending projections of subplate neurons into cortical layer 1, this molecule may be involved in the modulation of synaptic input to apical dendrites of pyramidal neurons.
Collapse
Affiliation(s)
- H Heuer
- Max-Planck Institute for Experimental Endocrinology, Feodor-Lynen-Str. 7, D-30625 Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
50
|
Hirano S, Bless D, Heisey D, Ford C. Roles of hepatocyte growth factor and transforming growth factor beta1 in production of extracellular matrix by canine vocal fold fibroblasts. Laryngoscope 2003; 113:144-8. [PMID: 12514399 DOI: 10.1097/00005537-200301000-00027] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND/OBJECTIVES When the lamina propria of the vocal fold is replaced by fibrosis after wound healing, it is difficult to restore an appropriate viscoelasticity of the vocal fold. To treat fibrotic scarring, material that reduces collagen deposition and increases soft amorphous substances, such as hyaluronic acid, is required. The potential use of hepatocyte growth factor (HGF) is intriguing. In this study, the authors examined canine vocal fold fibroblasts to determine how HGF contributes to the production of extracellular matrix. More specifically, the authors describe how the productions of hyaluronic acid, collagen type I, and fibronectin are associated with administration of HGF and transforming growth factor beta1. STUDY DESIGN In vitro. METHODS Fibroblasts were collected from the lamina propria of the vocal folds of five Beagles and were cultured with and without HGF or transforming growth factor beta1. The productions of hyaluronic acid, collagen type I, and fibronectin in supernatants culture were examined using ELISA. RESULTS Hepatocyte growth factor stimulated hyaluronic acid production, reduced collagen type I production, and did not affect fibronectin production, while transforming growth factor beta1 stimulated the productions of all components. CONCLUSIONS Collagen type I appears to be a major contributor in creation of fibrosis, and excessive fibronectin may stiffen the tissue. Since HGF reduced collagen type I production from fibroblasts and increased hyaluronic acid, HGF is considered to have therapeutic potential in prevention and treatment of the fibrosis of the vocal fold.
Collapse
Affiliation(s)
- Shigeru Hirano
- Department of Surgery, University of Wisconsin-Madison, 53792, USA.
| | | | | | | |
Collapse
|