1
|
Georgelou K, Saridaki EA, Karali K, Papagiannaki A, Charalampopoulos I, Gravanis A, Tzeranis DS. Microneurotrophin BNN27 Reduces Astrogliosis and Increases Density of Neurons and Implanted Neural Stem Cell-Derived Cells after Spinal Cord Injury. Biomedicines 2023; 11:biomedicines11041170. [PMID: 37189788 DOI: 10.3390/biomedicines11041170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/02/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Microneurotrophins, small-molecule mimetics of endogenous neurotrophins, have demonstrated significant therapeutic effects on various animal models of neurological diseases. Nevertheless, their effects on central nervous system injuries remain unknown. Herein, we evaluate the effects of microneurotrophin BNN27, an NGF analog, in the mouse dorsal column crush spinal cord injury (SCI) model. BNN27 was delivered systemically either by itself or combined with neural stem cell (NSC)-seeded collagen-based scaffold grafts, demonstrated recently to improve locomotion performance in the same SCI model. Data validate the ability of NSC-seeded grafts to enhance locomotion recovery, neuronal cell integration with surrounding tissues, axonal elongation and angiogenesis. Our findings also show that systemic administration of BNN27 significantly reduced astrogliosis and increased neuron density in mice SCI lesion sites at 12 weeks post injury. Furthermore, when BNN27 administration was combined with NSC-seeded PCS grafts, BNN27 increased the density of survived implanted NSC-derived cells, possibly addressing a major challenge of NSC-based SCI treatments. In conclusion, this study provides evidence that small-molecule mimetics of endogenous neurotrophins can contribute to effective combinatorial treatments for SCI, by simultaneously regulating key events of SCI and supporting grafted cell therapies in the lesion site.
Collapse
Affiliation(s)
- Konstantina Georgelou
- Department of Pharmacology, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 71003 Heraklion, Greece
| | | | - Kanelina Karali
- Department of Pharmacology, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 71003 Heraklion, Greece
| | - Argyri Papagiannaki
- Department of Pharmacology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 71003 Heraklion, Greece
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 71003 Heraklion, Greece
| | - Dimitrios S Tzeranis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 71003 Heraklion, Greece
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 2109, Cyprus
| |
Collapse
|
2
|
Zheng B, Tuszynski MH. Regulation of axonal regeneration after mammalian spinal cord injury. Nat Rev Mol Cell Biol 2023; 24:396-413. [PMID: 36604586 DOI: 10.1038/s41580-022-00562-y] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 01/06/2023]
Abstract
One hundred years ago, Ramón y Cajal, considered by many as the founder of modern neuroscience, stated that neurons of the adult central nervous system (CNS) are incapable of regenerating. Yet, recent years have seen a tremendous expansion of knowledge in the molecular control of axon regeneration after CNS injury. We now understand that regeneration in the adult CNS is limited by (1) a failure to form cellular or molecular substrates for axon attachment and elongation through the lesion site; (2) environmental factors, including inhibitors of axon growth associated with myelin and the extracellular matrix; (3) astrocyte responses, which can both limit and support axon growth; and (4) intraneuronal mechanisms controlling the establishment of an active cellular growth programme. We discuss these topics together with newly emerging hypotheses, including the surprising finding from transcriptomic analyses of the corticospinal system in mice that neurons revert to an embryonic state after spinal cord injury, which can be sustained to promote regeneration with neural stem cell transplantation. These gains in knowledge are steadily advancing efforts to develop effective treatment strategies for spinal cord injury in humans.
Collapse
Affiliation(s)
- Binhai Zheng
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA. .,VA San Diego Research Service, San Diego, CA, USA.
| | - Mark H Tuszynski
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA. .,VA San Diego Research Service, San Diego, CA, USA.
| |
Collapse
|
3
|
Hall A, Fortino T, Spruance V, Niceforo A, Harrop JS, Phelps PE, Priest CA, Zholudeva LV, Lane MA. Cell transplantation to repair the injured spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:79-158. [PMID: 36424097 PMCID: PMC10008620 DOI: 10.1016/bs.irn.2022.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Adam Hall
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Tara Fortino
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Victoria Spruance
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Division of Kidney, Urologic, & Hematologic Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alessia Niceforo
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - James S Harrop
- Department of Neurological and Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patricia E Phelps
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, United States
| | | | - Lyandysha V Zholudeva
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Gladstone Institutes, San Francisco, CA, United States
| | - Michael A Lane
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
4
|
Chiang MC, Chern E. Current Development, Obstacle and Futural Direction of Induced Pluripotent Stem Cell and Mesenchymal Stem Cell Treatment in Degenerative Retinal Disease. Int J Mol Sci 2022; 23:ijms23052529. [PMID: 35269671 PMCID: PMC8910526 DOI: 10.3390/ijms23052529] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 11/26/2022] Open
Abstract
Degenerative retinal disease is one of the major causes of vision loss around the world. The past several decades have witnessed emerging development of stem cell treatment for retinal disease. Nevertheless, sourcing stem cells remains controversial due to ethical concerns and their rarity. Furthermore, induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs) are both isolated from patients’ mature tissues; thus, issues such as avoiding moral controversy and adverse events related to immunosuppression and obtaining a large number of cells have opened a new era in regenerative medicine. This review focuses on the current application and development, clinical trials, and latest research of stem cell therapy, as well as its limitations and future directions.
Collapse
|
5
|
Kaplan B, Levenberg S. The Role of Biomaterials in Peripheral Nerve and Spinal Cord Injury: A Review. Int J Mol Sci 2022; 23:ijms23031244. [PMID: 35163168 PMCID: PMC8835501 DOI: 10.3390/ijms23031244] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/18/2022] Open
Abstract
Peripheral nerve and spinal cord injuries are potentially devastating traumatic conditions with major consequences for patients’ lives. Severe cases of these conditions are currently incurable. In both the peripheral nerves and the spinal cord, disruption and degeneration of axons is the main cause of neurological deficits. Biomaterials offer experimental solutions to improve these conditions. They can be engineered as scaffolds that mimic the nerve tissue extracellular matrix and, upon implantation, encourage axonal regeneration. Furthermore, biomaterial scaffolds can be designed to deliver therapeutic agents to the lesion site. This article presents the principles and recent advances in the use of biomaterials for axonal regeneration and nervous system repair.
Collapse
Affiliation(s)
- Ben Kaplan
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel;
- Bruce Rapaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel;
- Correspondence:
| |
Collapse
|
6
|
Begenisic T, Pavese C, Aiachini B, Nardone A, Rossi D. Dynamics of biomarkers across the stages of traumatic spinal cord injury - implications for neural plasticity and repair. Restor Neurol Neurosci 2021; 39:339-366. [PMID: 34657853 DOI: 10.3233/rnn-211169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Traumatic spinal cord injury (SCI) is a complex medical condition causing significant physical disability and psychological distress. While the adult spinal cord is characterized by poor regenerative potential, some recovery of neurological function is still possible through activation of neural plasticity mechanisms. We still have limited knowledge about the activation of these mechanisms in the different stages after human SCI. OBJECTIVE In this review, we discuss the potential role of biomarkers of SCI as indicators of the plasticity mechanisms at work during the different phases of SCI. METHODS An extensive review of literature related to SCI pathophysiology, neural plasticity and humoral biomarkers was conducted by consulting the PubMed database. Research and review articles from SCI animal models and SCI clinical trials published in English until January 2021 were reviewed. The selection of candidates for humoral biomarkers of plasticity after SCI was based on the following criteria: 1) strong evidence supporting involvement in neural plasticity (mandatory); 2) evidence supporting altered expression after SCI (optional). RESULTS Based on selected findings, we identified two main groups of potential humoral biomarkers of neural plasticity after SCI: 1) neurotrophic factors including: Brain derived neurotrophic factor (BDNF), Nerve growth factor (NGF), Neurotrofin-3 (NT-3), and Insulin-like growth factor 1 (IGF-1); 2) other factors including: Tumor necrosis factor-alpha (TNF-α), Matrix Metalloproteinases (MMPs), and MicroRNAs (miRNAs). Plasticity changes associated with these biomarkers often can be both adaptive (promoting functional improvement) and maladaptive. This dual role seems to be influenced by their concentrations and time-window during SCI. CONCLUSIONS Further studies of dynamics of biomarkers across the stages of SCI are necessary to elucidate the way in which they reflect the remodeling of neural pathways. A better knowledge about the mechanisms underlying plasticity could guide the selection of more appropriate therapeutic strategies to enhance positive spinal network reorganization.
Collapse
Affiliation(s)
- Tatjana Begenisic
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Chiara Pavese
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.,Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Beatrice Aiachini
- Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Antonio Nardone
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.,Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| |
Collapse
|
7
|
Sahib S, Sharma A, Menon PK, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Bryukhovetskiy I, Tian ZR, Patnaik R, Buzoianu AD, Wiklund L, Sharma HS. Cerebrolysin enhances spinal cord conduction and reduces blood-spinal cord barrier breakdown, edema formation, immediate early gene expression and cord pathology after injury. PROGRESS IN BRAIN RESEARCH 2020; 258:397-438. [PMID: 33223040 DOI: 10.1016/bs.pbr.2020.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Spinal cord evoked potentials (SCEP) are good indicators of spinal cord function in health and disease. Disturbances in SCEP amplitudes and latencies during spinal cord monitoring predict spinal cord pathology following trauma. Treatment with neuroprotective agents preserves SCEP and reduces cord pathology after injury. The possibility that cerebrolysin, a balanced composition of neurotrophic factors improves spinal cord conduction, attenuates blood-spinal cord barrier (BSCB) disruption, edema formation, and cord pathology was examined in spinal cord injury (SCI). SCEP is recorded from epidural space over rat spinal cord T9 and T12 segments after peripheral nerves stimulation. SCEP consists of a small positive peak (MPP), followed by a prominent negative peak (MNP) that is stable before SCI. A longitudinal incision (2mm deep and 5mm long) into the right dorsal horn (T10 and T11 segments) resulted in an immediate long-lasting depression of the rostral MNP with an increase in the latencies. Pretreatment with either cerebrolysin (CBL 5mL/kg, i.v. 30min before) alone or TiO2 nanowired delivery of cerebrolysin (NWCBL 2.5mL/kg, i.v.) prevented the loss of MNP amplitude and even enhanced further from the pre-injury level after SCI without affecting latencies. At 5h, SCI induced edema, BSCB breakdown, and cell injuries were significantly reduced by CBL and NWCBL pretreatment. Interestingly this effect on SCEP and cord pathology was still prominent when the NWCBL was delivered 2min after SCI. Moreover, expressions of c-fos and c-jun genes that are prominent at 5h in untreated SCI are also considerably reduced by CBL and NWCBL treatment. These results are the first to show that CBL and NWCBL enhanced SCEP activity and thwarted the development of cord pathology after SCI. Furthermore, NWCBL in low doses has superior neuroprotective effects on SCEP and cord pathology, not reported earlier. The functional significance and future clinical potential of CBL and NWCBL in SCI are discussed.
Collapse
Affiliation(s)
- Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Preeti K Menon
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden; Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
Lien BV, Brown NJ, Ransom SC, Lehrich BM, Shahrestani S, Tafreshi AR, Ransom RC, Sahyouni R. Enhancing peripheral nerve regeneration with neurotrophic factors and bioengineered scaffolds: A basic science and clinical perspective. J Peripher Nerv Syst 2020; 25:320-334. [DOI: 10.1111/jns.12414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Brian V. Lien
- School of Medicine University of California Irvine California USA
| | - Nolan J. Brown
- School of Medicine University of California Irvine California USA
| | - Seth C. Ransom
- College of Medicine University of Arkansas for Medical Sciences Little Rock Arkansas USA
| | - Brandon M. Lehrich
- Department of Biomedical Engineering University of California Irvine California USA
| | - Shane Shahrestani
- Keck School of Medicine University of Southern California Los Angeles California USA
- Department of Medical Engineering California Institute of Technology Pasadena California USA
| | - Ali R. Tafreshi
- Department of Neurological Surgery Geisinger Health System Danville Pennsylvania USA
| | - Ryan C. Ransom
- Department of Neurological Surgery Mayo Clinic Rochester Minnesota USA
| | - Ronald Sahyouni
- Department of Neurological Surgery University of California San Diego California USA
| |
Collapse
|
9
|
Dravid A, Parittotokkaporn S, Aqrawe Z, O’Carroll SJ, Svirskis D. Determining Neurotrophin Gradients in Vitro To Direct Axonal Outgrowth Following Spinal Cord Injury. ACS Chem Neurosci 2020; 11:121-132. [PMID: 31825204 DOI: 10.1021/acschemneuro.9b00565] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A spinal cord injury can damage neuronal connections required for both motor and sensory function. Barriers to regeneration within the central nervous system, including an absence of neurotrophic stimulation, impair the ability of injured neurons to reestablish their original circuitry. Exogenous neurotrophin administration has been shown to promote axonal regeneration and outgrowth following injury. The neurotrophins possess chemotrophic properties that guide axons toward the region of highest concentration. These growth factors have demonstrated potential to be used as a therapeutic intervention for orienting axonal growth beyond the injury lesion, toward denervated targets. However, the success of this approach is dependent on the appropriate spatiotemporal distribution of these molecules to ensure detection and navigation by the axonal growth cone. A number of in vitro gradient-based assays have been employed to investigate axonal response to neurotrophic gradients. Such platforms have helped elucidate the potential of applying a concentration gradient of neurotrophins to promote directed axonal regeneration toward a functionally significant target. Here, we review these techniques and the principles of gradient detection in axonal guidance, with particular focus on the use of neurotrophins to orient the trajectory of regenerating axons.
Collapse
Affiliation(s)
- Anusha Dravid
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Sam Parittotokkaporn
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Zaid Aqrawe
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Simon J. O’Carroll
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| |
Collapse
|
10
|
Toll-like receptor 9 antagonism modulates astrocyte function and preserves proximal axons following spinal cord injury. Brain Behav Immun 2019; 80:328-343. [PMID: 30953770 DOI: 10.1016/j.bbi.2019.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 03/14/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023] Open
Abstract
Increasing evidence indicates that innate immune receptors play important, yet controversial, roles in traumatic central nervous system (CNS) injury. Despite many advances, the contributions of toll-like receptors (TLRs) to spinal cord injury (SCI) remain inadequately defined. We previously reported that a toll-like receptor 9 (TLR9) antagonist, oligodeoxynucleotide 2088 (ODN 2088), administered intrathecally, improves the functional and histopathological outcomes of SCI. However, the molecular and cellular changes that occur at the injury epicenter following ODN 2088 treatment are not completely understood. Following traumatic SCI, a glial scar, consisting primarily of proliferating reactive astrocytes, forms at the injury epicenter and assumes both beneficial and detrimental roles. Increased production of chondroitin sulfate proteoglycans (CSPGs) by reactive astrocytes inhibits the regeneration of injured axons. Astrocytes express TLR9, which can be activated by endogenous ligands released by damaged cells. It is not yet known how TLR9 antagonism modifies astrocyte function at the glial scar and how this affects axonal preservation or re-growth following SCI. The present studies were undertaken to address these issues. We report that in female mice sustaining a severe mid-thoracic (T8) contusion injury, the number of proliferating astrocytes in regions rostral and caudal to the lesion border increased significantly by 30- and 24-fold, respectively, compared to uninjured controls. Intrathecal ODN 2088 treatment significantly reduced the number of proliferating astrocytes by 60% in both regions. This effect appeared to be, at least partly, mediated through the direct actions of ODN 2088 on astrocytes, since the antagonist decreased proliferation in pure SC astrocyte cultures by preventing the activation of the Erk/MAPK signaling pathway. In addition, CSPG immunoreactivity at the lesion border was more pronounced in vehicle-treated injured mice compared to uninjured controls and was significantly reduced following administration of ODN 2088 to injured mice. Moreover, ODN 2088 significantly decreased astrocyte migration in an in vitro scratch-wound assay. Anterograde tracing and quantification of corticospinal tract (CST) axons in injured mice, indicated that ODN 2088 preserves proximal axons. Taken together, these findings suggest that ODN 2088 modifies the glial scar and creates a milieu that fosters axonal protection at the injury site.
Collapse
|
11
|
Mesentier-Louro LA, Rosso P, Carito V, Mendez-Otero R, Santiago MF, Rama P, Lambiase A, Tirassa P. Nerve Growth Factor Role on Retinal Ganglion Cell Survival and Axon Regrowth: Effects of Ocular Administration in Experimental Model of Optic Nerve Injury. Mol Neurobiol 2019; 56:1056-1069. [PMID: 29869196 DOI: 10.1007/s12035-018-1154-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/24/2018] [Indexed: 01/04/2023]
Abstract
Retinal ganglion cell (RGC) degeneration occurs within 2 weeks following optic nerve crush (ONC) as a consequence of reduced retro-transport of growth factors including nerve growth factor (NGF). The hypothesis that intravitreal (ivt) and eye drop (ed) administration of recombinant human NGF (rhNGF) might counteract ONC in adult rats is explored in this study. We found that both ivt- and ed-rhNGF reduced RGC loss and stimulated axonal regrowth. Chiefly, survival and regenerative effects of rhNGF were associated with a reduction of cells co-expressing Nogo-A/p75NTR at crush site borders, which contribute to glia scar formation following nerve injury, and induce further degeneration. We also found that ocular application of rhNGF reduced p75NTR and proNGF and enhanced phosphorylation of TrkA and its intracellular signals at retina level. Nogo-R and Rock2 expression was also normalized by ed-rhNGF treatment in both ONC and contralateral retina. Our findings that ocular applied NGF reaches and exerts biological actions on posterior segment of the eye give a further insight into the neurotrophin diffusion/transport through eye structures and/or their trafficking in optic nerve. In addition, the use of a highly purified NGF form in injury condition in which proNGF/p75NTR binding is favored indicates that increased availability of mature NGF restores the balance between TrkA and p75NGF, thus resulting in RGC survival and axonal growth. In conclusion, ocular applied NGF is confirmed as a good experimental paradigm to study mechanisms of neurodegeneration and regeneration, disclose biomarkers, and time windows for efficacy treatment following cell or nerve injury.
Collapse
Affiliation(s)
- Louise A Mesentier-Louro
- Eye Repair Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pamela Rosso
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Via di Fosso di Fiorano, 64 (00143), Rome, Italy
| | - Valentina Carito
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Via di Fosso di Fiorano, 64 (00143), Rome, Italy
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo F Santiago
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paolo Rama
- Eye Repair Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Lambiase
- Section of Ophthalmology, Department of Sense Organs, University Sapienza, Rome, Italy
| | - Paola Tirassa
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Via di Fosso di Fiorano, 64 (00143), Rome, Italy.
| |
Collapse
|
12
|
Jin Y, Shumsky JS, Fischer I. Axonal regeneration of different tracts following transplants of human glial restricted progenitors into the injured spinal cord in rats. Brain Res 2018; 1686:101-112. [PMID: 29408659 DOI: 10.1016/j.brainres.2018.01.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/18/2018] [Accepted: 01/21/2018] [Indexed: 12/15/2022]
Abstract
The goal of this study was to compare the efficacy of human glial restricted progenitors (hGRPs) in promoting axonal growth of different tracts. We examined the potential of hGRPs grafted into a cervical (C4) dorsal column lesion to test sensory axons, and into a C4 hemisection to test motor tracts. The hGRPs, thawed from frozen stocks, were suspended in a PureCol matrix and grafted acutely into a C4 dorsal column or hemisection lesion. Control rats received PureCol only. Five weeks after transplantation, all transplanted cells survived in rats with the dorsal column lesion but only about half of the grafts in the hemisection. In the dorsal column lesion group, few sensory axons grew short distances into the lesion site of control animals. The presence of hGRPs transplants enhanced axonal growth significantly farther into the transplants. In the hemisection group, coerulospinal axons extended similarly into both control and transplant groups with no enhancement by the presence of hGRPs. Rubrospinal axons did not grow into the lesion even in the presence of hGRPs. However, reticulospinal and raphespinal axons grew for a significantly longer distance into the transplants. These results demonstrate the differential capacity of axonal growth/regeneration of the motor and sensory tracts based on their intrinsic abilities as well as their response to the modified environment induced by the hGRPs transplants. We conclude that hGRP transplants can modify the injury site for axon growth of sensory and some motor tracts, and suggest they could be combined with other interventions to restore connectivity.
Collapse
Affiliation(s)
- Ying Jin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| | - Jed S Shumsky
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| |
Collapse
|
13
|
Schinstine M, Fiore DM, Winn SR, Emerich DF. Polymer-Encapsulated Schwannoma Cells Expressing Human Nerve Growth Factor Promote the Survival of Cholinergic Neurons after a Fimbria-Fornix Transection. Cell Transplant 2017; 4:93-102. [PMID: 7728338 DOI: 10.1177/096368979500400113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Many investigators have recently used genetically modified primary fibroblasts of fibroblast cell lines (e.g., 3T3, 208F, or BHK cells) to deliver recombinant nerve growth factor (NGF) into the CNS. In the current study, SCT-1 cells, a Schwannoma cell line derived from a transgenic mouse, were transfected with a human NGF (hNGF) cDNA. After selection, these cells were encased within a polymer capsule and implanted into the ventricles of fimbria-fornix lesioned rats. Encapsulated, non-transfected cells served as controls. Results demonstrated that the hNGF transgene is expressed for at least 3 weeks after implantation. Moreover, the cells did not overgrow the capsule. Recombinant hNGF was able to save >70% of lesioned cholinergic neurons, as assessed by NGF-receptor (NGFr) and choline acetyltransferase (ChAT) immunohistochemistry, from cell death. The number of cholinergic neurons in animals that received control capsules (i.e., nontransfected SCT-1 cells) was similar to lesion only animals (i.e., ~27% and ~33% for NGFr- and ChAT-positive neurons, respectively. These results show that SCT-1 cells can be used to deliver biologically active hNGF into the lesioned rat brain.
Collapse
Affiliation(s)
- M Schinstine
- CytoTherapeutics, Inc., Dept. of Neuroscience, Providence, RI 02906, USA
| | | | | | | |
Collapse
|
14
|
Nakahara Y, Gage FH, Tuszynski MH. Grafts of Fibroblasts Genetically Modified to Secrete Ngf, Bdnf, Nt-3, or Basic Fgf Elicit Differential Responses in the Adult Spinal Cord. Cell Transplant 2017; 5:191-204. [PMID: 8689031 DOI: 10.1177/096368979600500209] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Neuronal and axonal responses to neurotrophic factors in the developing spinal cord have been relatively well characterized, but little is known about adult spinal responses to neurotrophic factors. We genetically modified primary rat fibroblasts to produce either nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), or basic fibroblast growth factor (bFGF), then grafted these neurotrophic factor-secreting cells into the central gray matter of the spinal cord in adult rats. Spinal cord lesions were not made prior to grafting. From 2 wk to 6 mo later, sensory neurites of dorsal root origin extensively penetrated NGF-, NT-3-, and bFGF-producing grafts, whereas BDNF-secreting grafts elicited no growth responses. Putative noradrenergic neurites also penetrated NGF-secreting cell grafts. Local motor and corticospinal motor axons did not penetrate any of the neurotrophic factor-secreting grafts. These results indicate that unlesioned or minimally lesioned adult spinal cord sensory and putative noradrenergic populations retain significant neurotrophic factor responsiveness, whereas motor neurites are comparatively resistant even to those neurotrophic factors to which they exhibit survival dependence during development. Grafts of genetically modified cells can be a useful tool for characterizing neurotrophic factor responsiveness in the adult spinal cord and designing strategies to promote axonal regeneration after injury.
Collapse
Affiliation(s)
- Y Nakahara
- Department of Neurosciences, University of California-San Diego, La Jolla 92093, USA
| | | | | |
Collapse
|
15
|
Tuszynski MH, Weidner N, McCormack M, Miller I, Powell H, Conner J. Grafts of Genetically Modified Schwann Cells to the Spinal Cord: Survival, Axon Growth, and Myelination. Cell Transplant 2017; 7:187-96. [PMID: 9588600 DOI: 10.1177/096368979800700213] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Schwann cells naturally support axonal regeneration after injury in the peripheral nervous system, and have also shown a significant, albeit limited, ability to support axonal growth and remyelination after grafting to the central nervous system (CNS). It is possible that Schwann cell-induced axonal growth in the CNS could be substantially increased by genetic manipulation to secrete augmented amounts of neurotrophic factors. To test this hypothesis, cultured primary adult rat Schwann cells were genetically modified using retroviral vectors to produce and secrete high levels of human nerve growth factor (NGF). These cells were then grafted to the midthoracic spinal cords of adult rats. Findings were compared to animals that received grafts of nontransduced Schwann cells. Spinal cord lesions were not placed prior to grafting because the primary aim of this study was to examine features of grafted Schwann cell survival, growth, and effects on host axons. In vitro prior to grafting, Schwann cells secreted 1.5 + 0.1 ng human NGF/ml/106 cells/day. Schwann cell transplants readily survived for 2 wk to 1 yr after in vivo placement. Some NGF-transduced grafts slowly increased in size over time compared to nontransduced grafts; the latter remained stable in size. NGF-transduced transplants were densely penetrated by primary sensory nociceptive axons originating from the dorsolateral fasciculus of the spinal cord, whereas control grafts showed significantly fewer penetrating sensory axons. Over time, Schwann cell grafts also became penetrated by TH- and DBH-labeled axons of putative coerulospinal origin, unlike control cell grafts. Ultrastructurally, axons in both graft types were extensively myelinated by Schwann cells. Grafted animals showed no changes in gross locomotor function. In vivo expression of the human NGF transgene was demonstrated for periods of at least 6 m. These findings demonstrate that primary adult Schwann cells 1) can be transduced to secrete augmented levels of neurotrophic factors, 2) survive grafting to the CNS for prolonged time periods, 3) elicit robust growth of host neurotrophin-responsive axons, 4) myelinate CNS axons, and 5) express the transgene for prolonged time periods in vivo. Some grafts slowly enlarge over time, a feature that may be attributable to the propensity of Schwann cells to immortalize after multiple passages. Transduced Schwann cells merit further study as tools for promoting CNS regeneration.
Collapse
Affiliation(s)
- M H Tuszynski
- Department of Neurosciences, University of California-San Diego, La Jolla 92093-0608, USA
| | | | | | | | | | | |
Collapse
|
16
|
Mizumoto H, Mizumoto K, Whiteley SJO, Shatos M, Klassen H, Young MJ. Transplantation of Human Neural Progenitor Cells to the Vitreous Cavity of the Royal College of Surgeons Rat. Cell Transplant 2017. [DOI: 10.3727/000000001783986936] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Hiroyuki Mizumoto
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Keiko Mizumoto
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Simon J. O. Whiteley
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Marie Shatos
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Henry Klassen
- CHOC Research, Children's Hospital of Orange County, Orange, CA 92868
| | - Michael J. Young
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
17
|
Blits B, Boer GJ, Verhaagen J. Pharmacological, Cell, and Gene Therapy Strategies to Promote Spinal Cord Regeneration. Cell Transplant 2017. [DOI: 10.3727/000000002783985521] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this review, recent studies using pharmacological treatment, cell transplantation, and gene therapy to promote regeneration of the injured spinal cord in animal models will be summarized. Pharmacological and cell transplantation treatments generally revealed some degree of effect on the regeneration of the injured ascending and descending tracts, but further improvements to achieve a more significant functional recovery are necessary. The use of gene therapy to promote repair of the injured nervous system is a relatively new concept. It is based on the development of methods for delivering therapeutic genes to neurons, glia cells, or nonneural cells. Direct in vivo gene transfer or gene transfer in combination with (neuro)transplantation (ex vivo gene transfer) appeared powerful strategies to promote neuronal survival and axonal regrowth following traumatic injury to the central nervous system. Recent advances in understanding the cellular and molecular mechanisms that govern neuronal survival and neurite outgrowth have enabled the design of experiments aimed at viral vector-mediated transfer of genes encoding neurotrophic factors, growth-associated proteins, cell adhesion molecules, and antiapoptotic genes. Central to the success of these approaches was the development of efficient, nontoxic vectors for gene delivery and the acquirement of the appropriate (genetically modified) cells for neurotransplantation. Direct gene transfer in the nervous system was first achieved with herpes viral and E1-deleted adenoviral vectors. Both vector systems are problematic in that these vectors elicit immunogenic and cytotoxic responses. Adeno-associated viral vectors and lentiviral vectors constitute improved gene delivery systems and are beginning to be applied in neuroregeneration research of the spinal cord. Ex vivo approaches were initially based on the implantation of genetically modified fibroblasts. More recently, transduced Schwann cells, genetically modified pieces of peripheral nerve, and olfactory ensheathing glia have been used as implants into the injured spinal cord.
Collapse
Affiliation(s)
- Bas Blits
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| | - Gerard J. Boer
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| | - Joost Verhaagen
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| |
Collapse
|
18
|
Tuszynski MH, Murai K, Blesch A, Grill R, Miller I. Functional Characterization of Ngf-Secreting Cell Grafts to the Acutely Injured Spinal Cord. Cell Transplant 2017; 6:361-8. [PMID: 9171168 DOI: 10.1177/096368979700600318] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Previously we reported that grafts of cells genetically modified to produce human nerve growth factor (hNGF) promoted specific and robust sprouting of spinal sensory, motor, and noradrenergic axons. In the present study we extend these investigations to assess NGF effects on corticospinal motor axons and on functional outcomes after spinal cord injury. Fibroblasts from adult rats were transduced to express human NGF; control cells were not genetically modified. Fibroblasts were then grafted to sites of midthoracic spinal cord dorsal hemisection lesions. Three months later, recipients of NGF-secreting grafts showed deficits on conditioned locomotion over a wire mesh that did not differ in extent from control-lesioned animals. On histological examination, NGF-secreting grafts elicited specific sprouting from spinal primary sensory afferent axons, local motor axons, and putative cerulospinal axons as previously reported, but no specific responses from corticospinal axons. Axons responding to NGF robustly penetrated the grafts but did not exit the grafts to extend to normal innervation territories distal to grafts. Grafted cells continued to express NGF protein through the experimental period of the study. These findings indicate that 1) spinal cord axons show directionally sensitive growth responses to neurotrophic factors, 2) growth of axons responding to a neurotrophic factor beyond an injury site and back to their natural target regions will likely require delivery of concentration gradients of neurotrophic factors toward the target, 3) corticospinal axons do not grow toward a cellular source of NGF, and 4) functional impairments are not improved by strictly local sprouting response of nonmotor systems.
Collapse
Affiliation(s)
- M H Tuszynski
- Department of Neurosciences, University of California-San Diego, La Jolla 92093-0608, USA
| | | | | | | | | |
Collapse
|
19
|
Tan J, Lu Z, Miao Z, Lei D, Zheng L, Zhao J. Effect of NGF From Venom of Chinese Cobra (Naja Atra)on Chondrocytes Proliferation and Metabolism In Vitro. J Cell Biochem 2017; 118:4308-4316. [DOI: 10.1002/jcb.26083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/19/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Jiachang Tan
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Key Laboratory of Regenerative MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- The Medical and Scientific Research CenterGuangxi Medical UniversityNanning530021China
| | - Zhenhui Lu
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Key Laboratory of Regenerative MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
| | - Zhikang Miao
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Key Laboratory of Regenerative MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
| | - Danqing Lei
- Department of Orthopaedics Trauma and Hand SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Key Laboratory of Regenerative MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- Guangxi Key Laboratory of Regenerative MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanning530021China
- The Medical and Scientific Research CenterGuangxi Medical UniversityNanning530021China
| |
Collapse
|
20
|
Lu Z, Lei D, Jiang T, Yang L, Zheng L, Zhao J. Nerve growth factor from Chinese cobra venom stimulates chondrogenic differentiation of mesenchymal stem cells. Cell Death Dis 2017; 8:e2801. [PMID: 28518137 PMCID: PMC5520725 DOI: 10.1038/cddis.2017.208] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/29/2017] [Accepted: 04/06/2017] [Indexed: 01/08/2023]
Abstract
Growth factors such as transforming growth factor beta1 (TGF-β1), have critical roles in the regulation of the chondrogenic differentiation of mesenchymal stem cells (MSCs), which promote cartilage repair. However, the clinical applications of the traditional growth factors are limited by their high cost, functional heterogeneity and unpredictable effects, such as cyst formation. It may be advantageous for cartilage regeneration to identify a low-cost substitute with greater chondral specificity and easy accessibility. As a neuropeptide, nerve growth factor (NGF) was involved in cartilage metabolism and NGF is hypothesized to mediate the chondrogenic differentiation of MSCs. We isolated NGF from Chinese cobra venom using a three-step procedure that we had improved upon from previous studies, and investigated the chondrogenic potential of NGF on bone marrow MSCs (BMSCs) both in vitro and in vivo. The results showed that NGF greatly upregulated the expression of cartilage-specific markers. When applied to cartilage repair for 4, 8 and 12 weeks, NGF-treated BMSCs have greater therapeutic effect than untreated BMSCs. Although inferior to TGF-β1 regarding its chondrogenic potential, NGF showed considerably lower expression of collagen type I, which is a fibrocartilage marker, and RUNX2, which is critical for terminal chondrocyte differentiation than TGF-β1, indicating its chondral specificity. Interestingly, NGF rarely induced BMSCs to differentiate into a neuronal phenotype, which may be due to the presence of other chondrogenic supplements. Furthermore, the underlying mechanism revealed that NGF-mediated chondrogenesis may be associated with the activation of PI3K/AKT and MAPK/ERK signaling pathways via the specific receptor of NGF, TrkA. In addition, NGF is easily accessed because of the abundance and low price of cobra venom, as well as the simplified methods for separation and purification. This study was the first to demonstrate the chondrogenic potential of NGF, which may provide a reference for cartilage regeneration in the clinic.
Collapse
Affiliation(s)
- Zhenhui Lu
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Danqing Lei
- The Medical and Scientific Research Center, Guangxi Medical University, Nanning, China
| | - Tongmeng Jiang
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lihui Yang
- School of Nursing, Guangxi Medical University, Nanning, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
21
|
Targeting Neurotrophins to Specific Populations of Neurons: NGF, BDNF, and NT-3 and Their Relevance for Treatment of Spinal Cord Injury. Int J Mol Sci 2017; 18:ijms18030548. [PMID: 28273811 PMCID: PMC5372564 DOI: 10.3390/ijms18030548] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Neurotrophins are a family of proteins that regulate neuronal survival, synaptic function, and neurotransmitter release, and elicit the plasticity and growth of axons within the adult central and peripheral nervous system. Since the 1950s, these factors have been extensively studied in traumatic injury models. Here we review several members of the classical family of neurotrophins, the receptors they bind to, and their contribution to axonal regeneration and sprouting of sensory and motor pathways after spinal cord injury (SCI). We focus on nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3), and their effects on populations of neurons within diverse spinal tracts. Understanding the cellular targets of neurotrophins and the responsiveness of specific neuronal populations will allow for the most efficient treatment strategies in the injured spinal cord.
Collapse
|
22
|
Nori S, Nakamura M, Okano H. Plasticity and regeneration in the injured spinal cord after cell transplantation therapy. PROGRESS IN BRAIN RESEARCH 2017; 231:33-56. [DOI: 10.1016/bs.pbr.2016.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Hodgetts SI, Harvey AR. Neurotrophic Factors Used to Treat Spinal Cord Injury. VITAMINS AND HORMONES 2016; 104:405-457. [PMID: 28215303 DOI: 10.1016/bs.vh.2016.11.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The application of neurotrophic factors as a therapy to improve morphological and behavioral outcomes after experimental spinal cord injury (SCI) has been the focus of many studies. These studies vary markedly in the type of neurotrophic factor that is delivered, the mode of administration, and the location, timing, and duration of the treatment. Generally, the majority of studies have had significant success if neurotrophic factors are applied in or close to the lesion site during the acute or the subacute phase after SCI. Comparatively fewer studies have administered neurotrophic factors in order to directly target the somata of injured neurons. The mode of delivery varies between acute injection of recombinant proteins, subacute or chronic delivery using a variety of strategies including osmotic minipumps, cell-mediated delivery, delivery using polymer release vehicles or supporting bridges of some sort, or the use of gene therapy to modify neurons, glial cells, or precursor/stem cells. In this brief review, we summarize the state of play of many of the therapies using these factors, most of which have been undertaken in rodent models of SCI.
Collapse
Affiliation(s)
- S I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia; Western Australian Neuroscience Research Institute, Perth, WA, Australia.
| | - A R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia; Western Australian Neuroscience Research Institute, Perth, WA, Australia
| |
Collapse
|
24
|
Singh S, Srivastava A, Srivastava P, Dhuriya YK, Pandey A, Kumar D, Rajpurohit CS. Advances in Stem Cell Research- A Ray of Hope in Better Diagnosis and Prognosis in Neurodegenerative Diseases. Front Mol Biosci 2016; 3:72. [PMID: 27878120 PMCID: PMC5099954 DOI: 10.3389/fmolb.2016.00072] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 10/24/2016] [Indexed: 12/13/2022] Open
Abstract
Neurodegeneration and neurodegenerative disorders have been a global health issue affecting the aging population worldwide. Recent advances in stem cell biology have changed the current face of neurodegenerative disease modeling, diagnosis, and transplantation therapeutics. Stem cells also serve the purpose of a simple in-vitro tool for screening therapeutic drugs and chemicals. We present the application of stem cells and induced pluripotent stem cells (iPSCs) in the field of neurodegeneration and address the issues of diagnosis, modeling, and therapeutic transplantation strategies for the most prevalent neurodegenerative disorders. We have discussed the progress made in the last decade and have largely focused on the various applications of stem cells in the neurodegenerative research arena.
Collapse
Affiliation(s)
- Shripriya Singh
- System Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology ResearchLucknow, India
- Academy of Scientific and Innovative ResearchLucknow, India
| | - Akriti Srivastava
- System Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology ResearchLucknow, India
| | - Pranay Srivastava
- System Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology ResearchLucknow, India
| | - Yogesh K. Dhuriya
- System Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology ResearchLucknow, India
- Academy of Scientific and Innovative ResearchLucknow, India
| | - Ankita Pandey
- System Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology ResearchLucknow, India
| | - Dipak Kumar
- System Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology ResearchLucknow, India
- Academy of Scientific and Innovative ResearchLucknow, India
| | - Chetan S. Rajpurohit
- System Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology ResearchLucknow, India
- Academy of Scientific and Innovative ResearchLucknow, India
| |
Collapse
|
25
|
Nothias JM, Mitsui T, Shumsky JS, Fischer I, Antonacci MD, Murray M. Combined Effects of Neurotrophin Secreting Transplants, Exercise, and Serotonergic Drug Challenge Improve Function in Spinal Rats. Neurorehabil Neural Repair 2016; 19:296-312. [PMID: 16263962 DOI: 10.1177/1545968305281209] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objectives. To determine the effects of neurotrophin-secreting transplants combined with exercise and serotonergic drug challenges on recovery of hindlimb function in rats with midthoracic spinal cord transection injuries. Methods. Spinalized animals received transplants of fibroblasts genetically modified to express brain-derived neurotrophic factor and neurotrophin-3 and daily cycling exercise. Hindlimb movement in an open-field test (BBB) was scored weekly. Serotonin agonists were used monthly to further stimulate motor function. Axonal growth was quantified in the transplant and at L5 using immunocytochemical markers. Weights of hindlimb muscles were used to assess muscle atrophy. Results. Neurotrophin-secreting transplants stimulated axonal growth, and cycling prevented muscle atrophy, but individual treatments did not improve motor scores. Combined treatments resulted in improvements in motor function. Serotonergic agonists further improved function in all groups, and transplant groups with exercise achieved weight-supporting levels following drug treatment. Conclusion. Combined treatments, but not individual treatments, improved hindlimb function.
Collapse
Affiliation(s)
- J-M Nothias
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | | | | | | | | | | |
Collapse
|
26
|
Tuszynski MH. Review : Gene Therapy: Applications to the Neurosciences and to Neurological Disease. Neuroscientist 2016. [DOI: 10.1177/107385849800400612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Gene expression is involved in some way in every human disease. As our knowledge of gene structure and function has blossomed in the last 2 decades, so too has our potential genetically-based repertoire for combating disease. Gene therapy refers to the manipulation of gene expression, either by augmenting the expression of therapeutic genes or by diminishing the expression of deleterious genes. In some neurological diseases, such as trauma, ischemia, and neurodegenerative disorders, gene therapy might be used to express genes for such substances as growth factors or neurotransmitters to prevent neuronal degeneration or to compensate for lost function, respectively. In other cases, gene therapy could be used to block the expression of genes that cause disease such as β-amyloid precursor protein or the Huntingtin gene. In inherited diseases of the nervous system such as muscular dystrophy, normal gene copies could be placed into the nervous system to compensate for lost function resulting from abnormal gene expression. The tools for achieving well-targeted, sustained, and safe gene delivery in the nervous system are now becoming available, and this technology is likely to substantially alter the nature of neurological therapy in the future. NEUROSCIENTIST 4:398-407, 1998
Collapse
Affiliation(s)
- Mark H. Tuszynski
- Department of Neurosciences University of California-San
Diego La Jolla, California Veterans Affairs Medical Center San Diego, California
| |
Collapse
|
27
|
Goganau I, Blesch A. Gene Therapy for Spinal Cord Injury. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
28
|
Sandner B, Ciatipis M, Motsch M, Soljanik I, Weidner N, Blesch A. Limited Functional Effects of Subacute Syngeneic Bone Marrow Stromal Cell Transplantation after Rat Spinal Cord Contusion Injury. Cell Transplant 2016; 25:125-39. [DOI: 10.3727/096368915x687679] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cell transplantation might be one means to improve motor, sensory, or autonomic recovery after traumatic spinal cord injury (SCI). Among the different cell types evaluated to date, bone marrow stromal cells (BMSCs) have received considerable interest due to their potential neuroprotective properties. However, uncertainty exists whether the efficacy of BMSCs after intraspinal transplantation justifies an invasive procedure. In the present study, we analyzed the effect of syngeneic BMSC transplantation following a moderate to severe rat spinal cord injury. Adult Fischer 344 rats underwent a T9 contusion injury (200 kDy) followed by grafting of GFP-expressing BMSCs 3 days postinjury. Animals receiving a contusion injury without cellular grafts or an injury followed by grafts of syngeneic GFP-expressing fibroblasts served as control. Eight weeks post-transplantation, BMSC-grafted animals showed only a minor effect in one measure of sensorimotor recovery, no significant differences in tissue sparing, and no changes in the recovery of bladder function compared to both control groups in urodynamic measurements. Both cell types survived in the lesion site with fibroblasts displaying a larger graft volume. Thus, contrary to some reports using allogeneic or xenogeneic transplants, subacute intraparenchymal grafting of syngeneic BMSCs has only a minor effect on functional recovery.
Collapse
Affiliation(s)
- Beatrice Sandner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Mareva Ciatipis
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie Motsch
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Irina Soljanik
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Armin Blesch
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
29
|
van Niekerk EA, Tuszynski MH, Lu P, Dulin JN. Molecular and Cellular Mechanisms of Axonal Regeneration After Spinal Cord Injury. Mol Cell Proteomics 2015; 15:394-408. [PMID: 26695766 DOI: 10.1074/mcp.r115.053751] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Indexed: 12/28/2022] Open
Abstract
Following axotomy, a complex temporal and spatial coordination of molecular events enables regeneration of the peripheral nerve. In contrast, multiple intrinsic and extrinsic factors contribute to the general failure of axonal regeneration in the central nervous system. In this review, we examine the current understanding of differences in protein expression and post-translational modifications, activation of signaling networks, and environmental cues that may underlie the divergent regenerative capacity of central and peripheral axons. We also highlight key experimental strategies to enhance axonal regeneration via modulation of intraneuronal signaling networks and the extracellular milieu. Finally, we explore potential applications of proteomics to fill gaps in the current understanding of molecular mechanisms underlying regeneration, and to provide insight into the development of more effective approaches to promote axonal regeneration following injury to the nervous system.
Collapse
Affiliation(s)
- Erna A van Niekerk
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093;
| | - Mark H Tuszynski
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093; §Veterans Administration Medical Center, San Diego, CA 92161
| | - Paul Lu
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093; §Veterans Administration Medical Center, San Diego, CA 92161
| | - Jennifer N Dulin
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093
| |
Collapse
|
30
|
Han ZM, Huang HM, Wang FF. Brain-derived neurotrophic factor gene-modified bone marrow mesenchymal stem cells. Exp Ther Med 2014; 9:519-522. [PMID: 25574226 PMCID: PMC4280929 DOI: 10.3892/etm.2014.2113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/29/2014] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to investigate the effects of human brain-derived neurotrophic factor (hBDNF) on the differentiation of bone marrow mesenchymal stem cells (MSCs) into neuron-like cells. Lentiviral vectors carrying the hBDNF gene were used to modify the bone marrow stromal cells (BMSCs) of Sprague-Dawley (SD) rats. The rat BMSCs were isolated, cultured and identified. A lentivirus bearing hBDNF and enhanced green fluorescent protein (eGFP) genes was subcultured and used to infect the SD rat BMSCs. The expression of eGFP was observed under a fluorescence microscope to determine the infection rate and growth of the transfected cells. Methylthiazolyldiphenyl-tetrazolium bromide (MTT) was used to detect the proliferation rate of cells following transfection. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis were used to detect the expression levels of hBDNF. Differentiation of neuron-like cells was induced in vitro and the differentiation rate of the induced neural-like cells was compared with that in control groups and analyzed statistically. In the cultured cells, flow cytometry demonstrated positive expression of cluster of differentiation (CD)90 and CD44, and negative expression of CD34 and CD45. The proliferation rate of the rat BMSCs increased following gene transfection. The expression of hBDNF-eGFP was detected in the BMSCs of the experimental group. The differentiation rate of hBDNF-modified cells into neuron-like cells in the experimental group was higher compared with that in empty plasmid and untransfected negative control groups. The difference was statistically significant (P<0.05). Thus, BDNF gene transfection is able to promote the differentiation of BMSCs into neuron-like cells. BDNF may play an important role in the differentiation of MSCs into neuron-like cells.
Collapse
Affiliation(s)
- Zhong-Min Han
- Department of Medical Technology, Zhengzhou Railway Vocational and Technical College, Zhengzhou, Henan 450052, P.R. China
| | - He-Mei Huang
- Department of Medical Technology, Zhengzhou Railway Vocational and Technical College, Zhengzhou, Henan 450052, P.R. China
| | - Fei-Fei Wang
- Department of Medical Technology, Zhengzhou Railway Vocational and Technical College, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
31
|
Harvey AR, Lovett SJ, Majda BT, Yoon JH, Wheeler LPG, Hodgetts SI. Neurotrophic factors for spinal cord repair: Which, where, how and when to apply, and for what period of time? Brain Res 2014; 1619:36-71. [PMID: 25451132 DOI: 10.1016/j.brainres.2014.10.049] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/20/2014] [Accepted: 10/23/2014] [Indexed: 12/22/2022]
Abstract
A variety of neurotrophic factors have been used in attempts to improve morphological and behavioural outcomes after experimental spinal cord injury (SCI). Here we review many of these factors, their cellular targets, and their therapeutic impact on spinal cord repair in different, primarily rodent, models of SCI. A majority of studies report favourable outcomes but results are by no means consistent, thus a major aim of this review is to consider how best to apply neurotrophic factors after SCI to optimize their therapeutic potential. In addition to which factors are chosen, many variables need be considered when delivering trophic support, including where and when to apply a given factor or factors, how such factors are administered, at what dose, and for how long. Overall, the majority of studies have applied neurotrophic support in or close to the spinal cord lesion site, in the acute or sub-acute phase (0-14 days post-injury). Far fewer chronic SCI studies have been undertaken. In addition, comparatively fewer studies have administered neurotrophic factors directly to the cell bodies of injured neurons; yet in other instructive rodent models of CNS injury, for example optic nerve crush or transection, therapies are targeted directly at the injured neurons themselves, the retinal ganglion cells. The mode of delivery of neurotrophic factors is also an important variable, whether delivered by acute injection of recombinant proteins, sub-acute or chronic delivery using osmotic minipumps, cell-mediated delivery, delivery using polymer release vehicles or supporting bridges of some sort, or the use of gene therapy to modify neurons, glial cells or precursor/stem cells. Neurotrophic factors are often used in combination with cell or tissue grafts and/or other pharmacotherapeutic agents. Finally, the dose and time-course of delivery of trophic support should ideally be tailored to suit specific biological requirements, whether they relate to neuronal survival, axonal sparing/sprouting, or the long-distance regeneration of axons ending in a different mode of growth associated with terminal arborization and renewed synaptogenesis. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Alan R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Sarah J Lovett
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Bernadette T Majda
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Jun H Yoon
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Lachlan P G Wheeler
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Stuart I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| |
Collapse
|
32
|
Aloe L, Bianchi P, De Bellis A, Soligo M, Rocco ML. Intranasal nerve growth factor bypasses the blood-brain barrier and affects spinal cord neurons in spinal cord injury. Neural Regen Res 2014; 9:1025-30. [PMID: 25206755 PMCID: PMC4146309 DOI: 10.4103/1673-5374.133161] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2014] [Indexed: 01/02/2023] Open
Abstract
The purpose of this work was to investigate whether, by intranasal administration, the nerve growth factor bypasses the blood-brain barrier and turns over the spinal cord neurons and if such therapeutic approach could be of value in the treatment of spinal cord injury. Adult Sprague-Dawley rats with intact and injured spinal cord received daily intranasal nerve growth factor administration in both nostrils for 1 day or for 3 consecutive weeks. We found an increased content of nerve growth factor and enhanced expression of nerve growth factor receptor in the spinal cord 24 hours after a single intranasal administration of nerve growth factor in healthy rats, while daily treatment for 3 weeks in a model of spinal cord injury improved the deficits in locomotor behaviour and increased spinal content of both nerve growth factor and nerve growth factor receptors. These outcomes suggest that the intranasal nerve growth factor bypasses blood-brain barrier and affects spinal cord neurons in spinal cord injury. They also suggest exploiting the possible therapeutic role of intranasally delivered nerve growth factor for the neuroprotection of damaged spinal nerve cells.
Collapse
Affiliation(s)
- Luigi Aloe
- Cellular Biology and Neurobiology Institute, National Research Council (CNR), Rome, Italy
| | - Patrizia Bianchi
- Cellular Biology and Neurobiology Institute, National Research Council (CNR), Rome, Italy
| | | | - Marzia Soligo
- Intitute of Translational Pharmacology, National Research Council (CNR), Rome, Italy
| | - Maria Luisa Rocco
- Cellular Biology and Neurobiology Institute, National Research Council (CNR), Rome, Italy
| |
Collapse
|
33
|
Amr SM, Gouda A, Koptan WT, Galal AA, Abdel-Fattah DS, Rashed LA, Atta HM, Abdel-Aziz MT. Bridging defects in chronic spinal cord injury using peripheral nerve grafts combined with a chitosan-laminin scaffold and enhancing regeneration through them by co-transplantation with bone-marrow-derived mesenchymal stem cells: case series of 14 patients. J Spinal Cord Med 2014; 37:54-71. [PMID: 24090088 PMCID: PMC4066552 DOI: 10.1179/2045772312y.0000000069] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE To investigate the effect of bridging defects in chronic spinal cord injury using peripheral nerve grafts combined with a chitosan-laminin scaffold and enhancing regeneration through them by co-transplantation with bone-marrow-derived mesenchymal stem cells. METHODS In 14 patients with chronic paraplegia caused by spinal cord injury, cord defects were grafted and stem cells injected into the whole construct and contained using a chitosan-laminin paste. Patients were evaluated using the International Standards for Classification of Spinal Cord Injuries. RESULTS Chitosan disintegration leading to post-operative seroma formation was a complication. Motor level improved four levels in 2 cases and two levels in 12 cases. Sensory-level improved six levels in two cases, five levels in five cases, four levels in three cases, and three levels in four cases. A four-level neurological improvement was recorded in 2 cases and a two-level neurological improvement occurred in 12 cases. The American Spinal Impairment Association (ASIA) impairment scale improved from A to C in 12 cases and from A to B in 2 cases. Although motor power improvement was recorded in the abdominal muscles (2 grades), hip flexors (3 grades), hip adductors (3 grades), knee extensors (2-3 grades), ankle dorsiflexors (1-2 grades), long toe extensors (1-2 grades), and plantar flexors (0-2 grades), this improvement was too low to enable them to stand erect and hold their knees extended while walking unaided. CONCLUSION Mesenchymal stem cell-derived neural stem cell-like cell transplantation enhances recovery in chronic spinal cord injuries with defects bridged by sural nerve grafts combined with a chitosan-laminin scaffold.
Collapse
Affiliation(s)
- Sherif M. Amr
- Department of Orthopaedics and Traumatology, Cairo University, Cairo, Egypt
| | - Ashraf Gouda
- Department of Orthopaedics and Traumatology, Al-Helal Hospital, Cairo, Egypt
| | - Wael T. Koptan
- Department of Orthopaedics and Traumatology, Cairo University, Cairo, Egypt
| | - Ahmad A. Galal
- Department of Orthopaedics and Traumatology, Cairo University, Cairo, Egypt
| | | | - Laila A. Rashed
- Department of Biochemistry and Molecular Biology, Cairo University, Cairo, Egypt
| | - Hazem M. Atta
- Department of Biochemistry and Molecular Biology, Cairo University, Cairo, Egypt
| | | |
Collapse
|
34
|
Abstract
A major challenge in repairing the injured spinal cord is to assure survival of damaged cells and to encourage regrowth of severed axons. Because neurotrophins are known to affect these processes during development, many experimental approaches to improving function of the injured spinal cord have made use of these agents, particularly Brain derived neurotrophic factor (BDNF) and Neurotrophin-3 (NT-3). More recently, neurotrophins have also been shown to affect the physiology of cells and synapses in the spinal cord. The effect of neurotrophins on circuit performance adds an important dimension to their consideration as agents for repairing the injured spinal cord. In this chapter we discuss the role of neurotrophins in promoting recovery after spinal cord injury from both a structural and functional perspective.
Collapse
Affiliation(s)
- Vanessa S Boyce
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | | |
Collapse
|
35
|
Silva NA, Sousa N, Reis RL, Salgado AJ. From basics to clinical: a comprehensive review on spinal cord injury. Prog Neurobiol 2013; 114:25-57. [PMID: 24269804 DOI: 10.1016/j.pneurobio.2013.11.002] [Citation(s) in RCA: 555] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 11/12/2013] [Accepted: 11/12/2013] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological disorder that affects thousands of individuals each year. Over the past decades an enormous progress has been made in our understanding of the molecular and cellular events generated by SCI, providing insights into crucial mechanisms that contribute to tissue damage and regenerative failure of injured neurons. Current treatment options for SCI include the use of high dose methylprednisolone, surgical interventions to stabilize and decompress the spinal cord, and rehabilitative care. Nonetheless, SCI is still a harmful condition for which there is yet no cure. Cellular, molecular, rehabilitative training and combinatorial therapies have shown promising results in animal models. Nevertheless, work remains to be done to ascertain whether any of these therapies can safely improve patient's condition after human SCI. This review provides an extensive overview of SCI research, as well as its clinical component. It starts covering areas from physiology and anatomy of the spinal cord, neuropathology of the SCI, current clinical options, neuronal plasticity after SCI, animal models and techniques to assess recovery, focusing the subsequent discussion on a variety of promising neuroprotective, cell-based and combinatorial therapeutic approaches that have recently moved, or are close, to clinical testing.
Collapse
Affiliation(s)
- Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Caldas das Taipas, Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
36
|
Enomoto M, Bunge MB, Tsoulfas P. A multifunctional neurotrophin with reduced affinity to p75NTR enhances transplanted Schwann cell survival and axon growth after spinal cord injury. Exp Neurol 2013; 248:170-82. [DOI: 10.1016/j.expneurol.2013.06.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/24/2013] [Accepted: 06/13/2013] [Indexed: 12/29/2022]
|
37
|
Zhang Y, Gao F, Wu D, Moshayedi P, Zhang X, Ellamushi H, Yeh J, Priestley JV, Bo X. Lentiviral mediated expression of a NGF-soluble Nogo receptor 1 fusion protein promotes axonal regeneration. Neurobiol Dis 2013; 58:270-80. [DOI: 10.1016/j.nbd.2013.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/14/2013] [Accepted: 06/19/2013] [Indexed: 10/26/2022] Open
|
38
|
Colello RJ, Chow WN, Bigbee JW, Lin C, Dalton D, Brown D, Jha BS, Mathern BE, Lee KD, Simpson DG. The incorporation of growth factor and chondroitinase ABC into an electrospun scaffold to promote axon regrowth following spinal cord injury. J Tissue Eng Regen Med 2013; 10:656-68. [DOI: 10.1002/term.1805] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 06/27/2013] [Accepted: 07/15/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Raymond J. Colello
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - Woon N. Chow
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - John W. Bigbee
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - Charles Lin
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - Dustin Dalton
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - Damien Brown
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - Balendu Shekhar Jha
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - Bruce E. Mathern
- Department of Neurosurgery, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - Kangmin D. Lee
- Department of Neurosurgery, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| | - David G. Simpson
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus; Virginia Commonwealth University; Richmond VA USA
| |
Collapse
|
39
|
Gao M, Lu P, Bednark B, Lynam D, Conner JM, Sakamoto J, Tuszynski MH. Templated agarose scaffolds for the support of motor axon regeneration into sites of complete spinal cord transection. Biomaterials 2012. [PMID: 23182350 DOI: 10.1016/j.biomaterials.2012.10.070] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bioengineered scaffolds have the potential to support and guide injured axons after spinal cord injury, contributing to neural repair. In previous studies we have reported that templated agarose scaffolds can be fabricated into precise linear arrays and implanted into the partially injured spinal cord, organizing growth and enhancing the distance over which local spinal cord axons and ascending sensory axons extend into a lesion site. However, most human injuries are severe, sparing only thin rims of spinal cord tissue in the margins of a lesion site. Accordingly, in the present study we examined whether template agarose scaffolds seeded with bone marrow stromal cells secreting Brain-Derived Neurotrophic Factor (BDNF) would support regeneration into severe, complete spinal cord transection sites. Moreover, we tested responses of motor axon populations originating from the brainstem. We find that templated agarose scaffolds support motor axon regeneration into a severe spinal cord injury model and organize axons into fascicles of highly linear configuration. BDNF significantly enhances axonal growth. Collectively, these findings support the feasibility of scaffold implantation for enhancing central regeneration after even severe central nervous system injury.
Collapse
Affiliation(s)
- Mingyong Gao
- Department of Neurosciences, University of California-San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Roet KCD, Eggers R, Verhaagen J. Noninvasive Bioluminescence Imaging of Olfactory Ensheathing Glia and Schwann Cells following Transplantation into the Lesioned Rat Spinal Cord. Cell Transplant 2012; 21:1853-65. [DOI: 10.3727/096368911x627471] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In this study, we assess the feasibility of bioluminescence imaging to monitor the survival of Schwann cells (SCs) and olfactory ensheathing glia cells (OECs) after implantation in the lesioned spinal cord of adult rats. To this end, purified SCs and OECs were genetically modified with lentiviral vectors encoding luciferase-2 and GFP and implanted in the lesioned dorsal column. The bioluminescent signal was monitored for over 3 months, and at 7 and 98 days postsurgery, the signal was compared to standard histological analysis of GFP expression in the spinal cords. The temporal profile of the bioluminescent signal showed three distinct phases for both cell types. (I) A relatively stable signal in the first week. (II) A progressive decline in signal strength in the second and third week. (III) After the third week, the average bioluminescent signal stabilized for both cell types. Interestingly, in the first week, the peak of the bioluminescent signal after luciferin injection was delayed when compared to later time points. Similar to in vitro, our data indicated a linear relationship between the in vivo bioluminescent signal and the GFP signal of the SCs and OECs in the spinal cords when the results of both the 7 and 98 day time points are combined. This is the first report of the use of in vivo bioluminescence to monitor cell survival in the lesioned rat spinal cord. Bioluminescence could be a potentially powerful, non-invasive strategy to examine the efficacy of treatments that aim to improve the survival of proregenerative cells transplanted in the injured rat spinal cord.
Collapse
Affiliation(s)
- Kasper C. D. Roet
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Ruben Eggers
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| |
Collapse
|
41
|
Tuszynski MH, Steward O. Concepts and methods for the study of axonal regeneration in the CNS. Neuron 2012; 74:777-91. [PMID: 22681683 PMCID: PMC3387806 DOI: 10.1016/j.neuron.2012.05.006] [Citation(s) in RCA: 227] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2012] [Indexed: 12/22/2022]
Abstract
Progress in the field of axonal regeneration research has been like the process of axonal growth itself: there is steady progress toward reaching the target, but there are episodes of mistargeting, misguidance along false routes, and connections that must later be withdrawn. This primer will address issues in the study of axonal growth after central nervous system injury in an attempt to provide guidance toward the goal of progress in the field. We address definitions of axonal growth, sprouting and regeneration after injury, and the research tools to assess growth.
Collapse
Affiliation(s)
- Mark H Tuszynski
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093-0662, USA.
| | | |
Collapse
|
42
|
Garbossa D, Boido M, Fontanella M, Fronda C, Ducati A, Vercelli A. Recent therapeutic strategies for spinal cord injury treatment: possible role of stem cells. Neurosurg Rev 2012; 35:293-311; discussion 311. [PMID: 22539011 DOI: 10.1007/s10143-012-0385-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 09/27/2011] [Accepted: 11/20/2011] [Indexed: 01/01/2023]
Abstract
Spinal cord injury (SCI) often results in significant dysfunction and disability. A series of treatments have been proposed to prevent and overcome the formation of the glial scar and inhibitory factors to axon regrowth. In the last decade, cell therapy has emerged as a new tool for several diseases of the nervous system. Stem cells act as minipumps providing trophic and immunomodulatory factors to enhance axonal growth, to modulate the environment, and to reduce neuroinflammation. This capability can be boosted by genetical manipulation to deliver trophic molecules. Different types of stem cells have been tested, according to their properties and the therapeutic aims. They differ from each other for origin, developmental stage, stage of differentiation, and fate lineage. Related to this, stem cells differentiating into neurons could be used for cell replacement, even though the feasibility that stem cells after transplantation in the adult lesioned spinal cord can differentiate into neurons, integrate within neural circuits, and emit axons reaching the muscle is quite remote. The timing of cell therapy has been variable, and may be summarized in the acute and chronic phases of disease, when stem cells interact with a completely different environment. Even though further experimental studies are needed to elucidate the mechanisms of action, the therapeutic, and the side effects of cell therapy, several clinical protocols have been tested or are under trial. Here, we report the state-of-the-art of cell therapy in SCI, in terms of feasibility, outcome, and side effects.
Collapse
Affiliation(s)
- D Garbossa
- Department of Neurosurgery, S. Giovanni Battista Hospital, University of Torino, Via Cherasco 15, 10126, Torino, Italy.
| | | | | | | | | | | |
Collapse
|
43
|
Nout YS, Rosenzweig ES, Brock JH, Strand SC, Moseanko R, Hawbecker S, Zdunowski S, Nielson JL, Roy RR, Courtine G, Ferguson AR, Edgerton VR, Beattie MS, Bresnahan JC, Tuszynski MH. Animal models of neurologic disorders: a nonhuman primate model of spinal cord injury. Neurotherapeutics 2012; 9:380-92. [PMID: 22427157 PMCID: PMC3337011 DOI: 10.1007/s13311-012-0114-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Primates are an important and unique animal resource. We have developed a nonhuman primate model of spinal cord injury (SCI) to expand our knowledge of normal primate motor function, to assess the impact of disease and injury on sensory and motor function, and to test candidate therapies before they are applied to human patients. The lesion model consists of a lateral spinal cord hemisection at the C7 spinal level with subsequent examination of behavioral, electrophysiological, and anatomical outcomes. Results to date have revealed significant neuroanatomical and functional differences between rodents and primates that impact the development of candidate therapies. Moreover, these findings suggest the importance of testing some therapeutic approaches in nonhuman primates prior to the use of invasive approaches in human clinical trials. Our primate model is intended to: 1) lend greater positive predictive value to human translatable therapies, 2) develop appropriate methods for human translation, 3) lead to basic discoveries that might not be identified in rodent models and are relevant to human translation, and 4) identify new avenues of basic research to "reverse-translate" important questions back to rodent models.
Collapse
Affiliation(s)
- Yvette S. Nout
- />Department of Animal and Veterinary Sciences, College of Agriculture, California State Polytechnic University, Pomona, CA USA
| | - Ephron S. Rosenzweig
- />Department of Neurosciences, University of California, La Jolla, San Diego, CA USA
| | - John H. Brock
- />Department of Neurosciences, University of California, La Jolla, San Diego, CA USA
| | - Sarah C. Strand
- />California National Primate Research Center, University of California, Davis, CA USA
| | - Rod Moseanko
- />California National Primate Research Center, University of California, Davis, CA USA
| | - Stephanie Hawbecker
- />California National Primate Research Center, University of California, Davis, CA USA
| | - Sharon Zdunowski
- />Department of Integrative Biology and Physiology, Los Angeles, CA USA
- />Brain Research Institute, University of California, Los Angeles, CA USA
| | - Jessica L. Nielson
- />Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, CA USA
| | - Roland R. Roy
- />Department of Integrative Biology and Physiology, Los Angeles, CA USA
- />Brain Research Institute, University of California, Los Angeles, CA USA
| | - Gregoire Courtine
- />Experimental Neurorehabilitation, Department of Neurology, Universität Zurich, Zurich, Switzerland
| | - Adam R. Ferguson
- />Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, CA USA
| | - V. Reggie Edgerton
- />Department of Integrative Biology and Physiology, Los Angeles, CA USA
- />Departments of Neurobiology and Neurosurgery, Los Angeles, CA USA
- />Brain Research Institute, University of California, Los Angeles, CA USA
| | - Michael S. Beattie
- />Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, CA USA
| | - Jacqueline C. Bresnahan
- />Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, CA USA
| | - Mark H. Tuszynski
- />Department of Neurosciences, University of California, La Jolla, San Diego, CA USA
- />Veterans Administration Medical Center, La Jolla, CA USA
| |
Collapse
|
44
|
McCreedy DA, Sakiyama-Elbert SE. Combination therapies in the CNS: engineering the environment. Neurosci Lett 2012; 519:115-21. [PMID: 22343313 DOI: 10.1016/j.neulet.2012.02.025] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 02/03/2012] [Accepted: 02/08/2012] [Indexed: 01/03/2023]
Abstract
The inhibitory extracellular environment that develops in response to traumatic brain injury and spinal cord injury hinders axon growth thereby limiting restoration of function. Several strategies have been developed to engineer a more permissive central nervous system (CNS) environment to promote regeneration and functional recovery. The multi-faced inhibitory nature of the CNS lesion suggests that therapies used in combination may be more effective. In this mini-review we summarize the most recent attempts to engineer the CNS extracellular environment after injury using combinatorial strategies. The advantages and limits of various combination therapies utilizing neurotrophin delivery, cell transplantation, and biomaterial scaffolds are discussed. Treatments that reduce the inhibition by chondroitin sulfate proteoglycans, myelin-associated inhibitors, and other barriers to axon regeneration are also reviewed. Based on the current state of the field, future directions are suggested for research on combination therapies in the CNS.
Collapse
Affiliation(s)
- Dylan A McCreedy
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Dr. Box 1097, St. Louis, MO 63130, United States
| | | |
Collapse
|
45
|
Abstract
Spinal cord injury (SCI) has multiple consequences, ranging from molecular imbalances to glial scar formation to functional impairments. It is logical to think that a combination of single treatments implemented in the right order and at the right time will be required to repair the spinal cord. However, the single treatments that compose the combination therapy will need to be chosen with caution as many have multiple outcomes that may or may not be synergistic. Single treatments may also elicit unwanted side-effects and/or effects that would decrease the repair potential of other components and/or the entire combination therapy. In this chapter a number of single treatments are discussed with respect to their multiplicity of action. These include strategies to boost growth and survival (such as neurotrophins and cyclic AMP) and strategies to reduce inhibitory factors (such as antimyelin-associated growth inhibitors and digestion of glial scar-associated inhibitors). We also present an overview of combination therapies that have successfully or unsuccessfully been tested in the laboratory using animal models. To effectively design a combination therapy a number of considerations need to be made such as the nature and timing of the treatments and the method for delivery. This chapter discusses these issues as well as considerations related to chronic SCI and the logistics of bringing combination therapies to the clinic.
Collapse
Affiliation(s)
- M Oudega
- Departments of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
46
|
Lee KB, Choi JH, Byun K, Chung KH, Ahn JH, Jeong GB, Hwang IK, Kim S, Won MH, Lee B. Recovery of CNS pathway innervating the sciatic nerve following transplantation of human neural stem cells in rat spinal cord injury. Cell Mol Neurobiol 2012; 32:149-57. [PMID: 21833549 PMCID: PMC11498461 DOI: 10.1007/s10571-011-9745-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 07/27/2011] [Indexed: 01/17/2023]
Abstract
Stem cell research has been attained a greater attention in most fields of medicine due to its potential for many incurable diseases through replacing or helping the regeneration of damaged cells or tissues. Here, we demonstrated the functional recovery and structural connection of the central nervous system pathway innervating the sciatic nerve after total transection of the spinal cord followed by the transplantation of human neural stem cells (hNSC) in the injured rat spinal cord site. The limb function of hNSC-treated group recovered dramatically compared with that in the sham group by Basso-Beattie-Bresnahan (BBB) scores. Transplanted hNSC differentiated into astrocytes and neurons in the injured site. In addition, immunohistochemistry for growth-associated protein 43 showed axonal regeneration in the injured spinal cord site. The pseudorabies viral-Ba (PRV-Ba) tracing method revealed that transplanted hNSC and their differentiated neurons showed positive labeling after sciatic nerve injection. In addition, the PRV-Ba labeling was also observed in several nuclei in the brain innervating the sciatic nerve. This result implies that the rat CNS motor pathway could be reconstructed by hNSC transplantation, and it may contribute to the functional recovery of the limb.
Collapse
Affiliation(s)
- Kwang-Bok Lee
- Department of Orthopedic Surgery, Chonbuk National University Medical School, Jeonju, 560-762 South Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon, 200-701 South Korea
| | - Kyunghee Byun
- Center for Genomics and Proteomics, Stem Cell Core Facility, Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Incheon, 406-840 South Korea
- Department of Anatomy and Cell Biology, Gachon University of Medicine and Science, Incheon, 406-799 South Korea
| | - Kwang Hoon Chung
- Department of Orthopedic Surgery, Jeju National University Medical School, Jeju, 690-767 South Korea
| | - Ji Hyeon Ahn
- Laboratory of Neuroscience, Department of Physical Therapy, College of Rehabilitation Science, Daegu University, Gyeongsan, 712-714 South Korea
| | - Goo-Bo Jeong
- Department of Anatomy and Cell Biology, Gachon University of Medicine and Science, Incheon, 406-799 South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Seungup Kim
- Department of Neurology, University of British Columbia, Vancouver, Canada
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701 South Korea
| | - Bonghee Lee
- Center for Genomics and Proteomics, Stem Cell Core Facility, Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Incheon, 406-840 South Korea
- Department of Anatomy and Cell Biology, Gachon University of Medicine and Science, Incheon, 406-799 South Korea
| |
Collapse
|
47
|
Bunge MB, Wood PM. Realizing the maximum potential of Schwann cells to promote recovery from spinal cord injury. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:523-540. [PMID: 23098734 DOI: 10.1016/b978-0-444-52137-8.00032-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Transplantation of Schwann cells (SCs) has been extensively investigated as a therapeutic intervention in rodent models of spinal cord injury (SCI). Here we review both strengths and weaknesses of this approach and discuss additional strategies for maximizing the potential of SCs to repair the injured spinal cord. With no additional treatments, SCs were consistently shown to provide a bridge across the lesion site, supporting the ingrowth of sensory and propriospinal axons, to myelinate axons and to decrease the size of cavities formed after injury. Supraspinal axons did not, however, grow onto the bridge, axons failed to traverse the caudal SC-host cord interface and transplanted SC survival was poor. More recent studies have shown that the potential of SC transplantation as a therapeutic approach can be strongly enhanced by combining additional strategies . For example, combining SC transplantation with elevation of cAMP levels resulted in growth of brainstem axons into the SC graft and caudal to the lesion and in significant improvements in locomotion. Axon growth (and functional improvement) have been increased by strategies to raise neurotrophin levels, either by injection or by genetic modification of the SCs before transplantation. A major problem in maximizing SC potential in injured cord has been in achieving good integration of the transplanted cells with the adjacent cord parenchyma. Several previous studies suggested an ability of SCs to migrate extensively in CNS tissue when astroctyes were absent and to myelinate CNS axons. Furthermore, in some cases involving very limited injury, SCs migrated and integrated well even in the presence of host astrocytes. Consistent with these observations, treatments with an enzyme, chondroitinase, to modify the SC-astrocyte interface surrounding the graft, have shown much promise. Very new studies have shown that SCs derived from SC precursors show a higher ability to survive, integrate well with host tissue and support brainstem axon growth into and beyond the graft, confirming the innate promise of SCs in spinal cord repair. We review one clinical trial already underway in Iran testing SC transplantation in patients with SCI. Finally, we briefly describe a protocol, adaptable to the principles of good manufacturing practice, for generating large numbers of human SCs. Overall, the available evidence suggests that SCs, especially when used in combination with other treatments, offer one of the best hopes we have today of devising an effective treatment for spinal cord repair.
Collapse
Affiliation(s)
- Mary Bartlett Bunge
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| | | |
Collapse
|
48
|
Gene therapy, neurotrophic factors and spinal cord regeneration. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:563-74. [PMID: 23098737 DOI: 10.1016/b978-0-444-52137-8.00035-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significant advances have been made in understanding the mechanisms that limit axon regeneration in the adult mammalian central nervous system and in addressing some of the obstacles for axon growth. Despite this progress numerous challenges remain to achieve regeneration of a large number of axons sufficient to mediate functional improvement. Given the complexity of injury-induced changes in axon, cell body, and parenchyma surrounding a spinal cord lesion, it seems likely that multiple factors both intrinsic and extrinsic to injured neurons have to be addressed to augment axon regeneration and useful reorganization of spared circuitry. Neurotrophic factors have been shown to be one potent means to increase the number and range of regenerating axons, to guide regenerating axons across a lesion site, and to augment regenerative cell body responses to injury. In this chapter we will review the potential and current limitations of neurotrophic factors and gene therapy, in combination with cellular transplants, for axon regeneration and sprouting in the injured spinal cord.
Collapse
|
49
|
Kitamura K, Fujiyoshi K, Yamane JI, Toyota F, Hikishima K, Nomura T, Funakoshi H, Nakamura T, Aoki M, Toyama Y, Okano H, Nakamura M. Human hepatocyte growth factor promotes functional recovery in primates after spinal cord injury. PLoS One 2011; 6:e27706. [PMID: 22140459 PMCID: PMC3226561 DOI: 10.1371/journal.pone.0027706] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 10/23/2011] [Indexed: 12/16/2022] Open
Abstract
Many therapeutic interventions for spinal cord injury (SCI) using neurotrophic factors have focused on reducing the area damaged by secondary, post-injury degeneration, to promote functional recovery. Hepatocyte growth factor (HGF), which is a potent mitogen for mature hepatocytes and a mediator of the inflammatory responses to tissue injury, was recently highlighted as a potent neurotrophic factor in the central nervous system. We previously reported that introducing exogenous HGF into the injured rodent spinal cord using a herpes simplex virus-1 vector significantly reduces the area of damaged tissue and promotes functional recovery. However, that study did not examine the therapeutic effects of administering HGF after injury, which is the most critical issue for clinical application. To translate this strategy to human treatment, we induced a contusive cervical SCI in the common marmoset, a primate, and then administered recombinant human HGF (rhHGF) intrathecally. Motor function was assessed using an original open field scoring system focusing on manual function, including reach-and-grasp performance and hand placement in walking. The intrathecal rhHGF preserved the corticospinal fibers and myelinated areas, thereby promoting functional recovery. In vivo magnetic resonance imaging showed significant preservation of the intact spinal cord parenchyma. rhHGF-treatment did not give rise to an abnormal outgrowth of calcitonin gene related peptide positive fibers compared to the control group, indicating that this treatment did not induce or exacerbate allodynia. This is the first study to report the efficacy of rhHGF for treating SCI in non-human primates. In addition, this is the first presentation of a novel scale for assessing neurological motor performance in non-human primates after contusive cervical SCI.
Collapse
Affiliation(s)
- Kazuya Kitamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Orthopedic Surgery, Hiratsuka City Hospital, Hiratsuka, Kanagawa, Japan
| | - Kanehiro Fujiyoshi
- Department of Orthopedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Jun-ichi Yamane
- Department of Orthopedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Fumika Toyota
- Central Institute for Experimental Animals, Miyamae-ku, Kawasaki, Kanagawa Japan
| | - Keigo Hikishima
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Central Institute for Experimental Animals, Miyamae-ku, Kawasaki, Kanagawa Japan
| | - Tatsuji Nomura
- Central Institute for Experimental Animals, Miyamae-ku, Kawasaki, Kanagawa Japan
| | - Hiroshi Funakoshi
- Center for Advanced Research and Education, Asahikawa Medical University, Midorigaoka, Asahikawa, Japan
| | - Toshikazu Nakamura
- Kringle Pharma Joint Research Division for Regenerative Drug Discovery, Center for Advanced Science and Innovation, Osaka University, Osaka, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Yoshiaki Toyama
- Department of Orthopedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
50
|
Kitamura K, Fujiyoshi K, Yamane JI, Toyota F, Hikishima K, Nomura T, Funakoshi H, Nakamura T, Aoki M, Toyama Y, Okano H, Nakamura M. Human hepatocyte growth factor promotes functional recovery in primates after spinal cord injury. PLoS One 2011. [PMID: 22140459 DOI: 10.1016/10.1371/journal.pone.0027706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Many therapeutic interventions for spinal cord injury (SCI) using neurotrophic factors have focused on reducing the area damaged by secondary, post-injury degeneration, to promote functional recovery. Hepatocyte growth factor (HGF), which is a potent mitogen for mature hepatocytes and a mediator of the inflammatory responses to tissue injury, was recently highlighted as a potent neurotrophic factor in the central nervous system. We previously reported that introducing exogenous HGF into the injured rodent spinal cord using a herpes simplex virus-1 vector significantly reduces the area of damaged tissue and promotes functional recovery. However, that study did not examine the therapeutic effects of administering HGF after injury, which is the most critical issue for clinical application. To translate this strategy to human treatment, we induced a contusive cervical SCI in the common marmoset, a primate, and then administered recombinant human HGF (rhHGF) intrathecally. Motor function was assessed using an original open field scoring system focusing on manual function, including reach-and-grasp performance and hand placement in walking. The intrathecal rhHGF preserved the corticospinal fibers and myelinated areas, thereby promoting functional recovery. In vivo magnetic resonance imaging showed significant preservation of the intact spinal cord parenchyma. rhHGF-treatment did not give rise to an abnormal outgrowth of calcitonin gene related peptide positive fibers compared to the control group, indicating that this treatment did not induce or exacerbate allodynia. This is the first study to report the efficacy of rhHGF for treating SCI in non-human primates. In addition, this is the first presentation of a novel scale for assessing neurological motor performance in non-human primates after contusive cervical SCI.
Collapse
Affiliation(s)
- Kazuya Kitamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|