51
|
Mukhamedshina YO, Garanina EE, Masgutova GA, Galieva LR, Sanatova ER, Chelyshev YA, Rizvanov AA. Assessment of Glial Scar, Tissue Sparing, Behavioral Recovery and Axonal Regeneration following Acute Transplantation of Genetically Modified Human Umbilical Cord Blood Cells in a Rat Model of Spinal Cord Contusion. PLoS One 2016; 11:e0151745. [PMID: 27003408 PMCID: PMC4803326 DOI: 10.1371/journal.pone.0151745] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 03/03/2016] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE AND METHODS This study investigated the potential for protective effects of human umbilical cord blood mononuclear cells (UCB-MCs) genetically modified with the VEGF and GNDF genes on contusion spinal cord injury (SCI) in rats. An adenoviral vector was constructed for targeted delivery of VEGF and GDNF to UCB-MCs. Using a rat contusion SCI model we examined the efficacy of the construct on tissue sparing, glial scar severity, the extent of axonal regeneration, recovery of motor function, and analyzed the expression of the recombinant genes VEGF and GNDF in vitro and in vivo. RESULTS Transplantation of UCB-MCs transduced with adenoviral vectors expressing VEGF and GDNF at the site of SCI induced tissue sparing, behavioral recovery and axonal regeneration comparing to the other constructs tested. The adenovirus encoding VEGF and GDNF for transduction of UCB-MCs was shown to be an effective and stable vehicle for these cells in vivo following the transplantation into the contused spinal cord. CONCLUSION Our results show that a gene delivery using UCB-MCs-expressing VEGF and GNDF genes improved both structural and functional parameters after SCI. Further histological and behavioral studies, especially at later time points, in animals with SCI after transplantation of genetically modified UCB-MCs (overexpressing VEGF and GDNF genes) will provide additional insight into therapeutic potential of such cells.
Collapse
Affiliation(s)
- Yana O. Mukhamedshina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Tatarstan, Kazan, Russia
| | - Ekaterina E. Garanina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Tatarstan, Kazan, Russia
| | - Galina A. Masgutova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Tatarstan, Kazan, Russia
| | - Luisa R. Galieva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Tatarstan, Kazan, Russia
| | - Elvira R. Sanatova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Tatarstan, Kazan, Russia
| | - Yurii A. Chelyshev
- Department of histology, Kazan State Medical University, Tatarstan, Kazan, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Tatarstan, Kazan, Russia
- * E-mail:
| |
Collapse
|
52
|
Zhao YZ, Jiang X, Xiao J, Lin Q, Yu WZ, Tian FR, Mao KL, Yang W, Wong HL, Lu CT. Using NGF heparin-poloxamer thermosensitive hydrogels to enhance the nerve regeneration for spinal cord injury. Acta Biomater 2016; 29:71-80. [PMID: 26472614 PMCID: PMC7517710 DOI: 10.1016/j.actbio.2015.10.014] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/22/2015] [Accepted: 10/09/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Nerve growth factor (NGF) has potential in spinal cord injury (SCI) therapy, but limited by the poor physicochemical stability and low ability to cross the blood spinal cord barrier. Novel heparin-poloxamer (HP) thermo-sensitive hydrogel was constructed to enhance the NGF regeneration on SCI. METHOD NGF-HP thermo-sensitive hydrogel was prepared and related characteristics including gelation temperature, rheological behavior and micromorphology were measured. Local NGF delivery to the injured spinal cord was achieved by in situ injection in the injured space. The cellular uptake of NGF-HP hydrogel was evaluated with PC12 cells in vitro. Pathologic characteristics and neuron regeneration effects on the SCI rats were studied to evaluate the enhanced therapy of NGF-HP hydrogel. Endoplasmic reticulum (ER) stress-induced apoptosis was analyzed to explore the related mechanism in SCI regeneration. RESULTS NGF-HP hydrogel showed good morphology and stable bioactivity of NGF in vitro. NGF-HP hydrogel combined treatment significantly enhanced the efficiency of NGF cellular uptake (P<0.05) without obvious cytotoxicity. Significant improvements in both neuron functions and tissue morphology on the SCI rats were observed in NGF-HP hydrogel group. Compared with free HP hydrogel and NGF treatment groups, NGF-HP hydrogel group showed significant inhibition on the formation of glial scars in the extreme crushed rat SCI model. The neuroprotective effects of NGF-HP were related to the inhibition of chronic ER stress-induced apoptosis. CONCLUSIONS HP hydrogel combined with orthotopic injection technique might be an effective method to deliver NGF into the injured site, which will provide an effective strategy for SCI regeneration. STATEMENT OF SIGNIFICANCE Spinal cord injury (SCI) is a devastating condition that can lead to sudden loss of sensory and autonomic function. Current treatment includes decompression surgery, injury stabilization, secondary complications prevention and rehabilitation. However, neurological recovery is limited. Nerve growth factor (NGF) has potential in SCI therapy, but limited by the poor physicochemical stability and low ability to cross the blood spinal cord barrier. Hydrogels have good affinity and compatibility to biological tissue. In this study, we developed a novel heparin-poloxamer (HP) thermo-sensitive hydrogel to enhance the spinal cord regeneration of NGF. From SCI rat experiment, HP hydrogel combined with orthotopic injection technique showed best neuroprotective effects among experimental groups. This novel combined technique will provide an effective strategy for SCI regeneration.
Collapse
Affiliation(s)
- Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Hainan Medical College, Haikou City, Hainan Province 571000, China
| | - Xi Jiang
- Zhejiang University Mingzhou Hospital, Zhejiang Province 315104, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Qian Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Wen-Ze Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Fu-Rong Tian
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Kai-Li Mao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Wei Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Ho Lun Wong
- School of Pharmacy, Temple University, Philadelphia, PA 19140, USA.
| | - Cui-Tao Lu
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| |
Collapse
|
53
|
Rat Nasal Respiratory Mucosa-Derived Ectomesenchymal Stem Cells Differentiate into Schwann-Like Cells Promoting the Differentiation of PC12 Cells and Forming Myelin In Vitro. Stem Cells Int 2015; 2015:328957. [PMID: 26339250 PMCID: PMC4539076 DOI: 10.1155/2015/328957] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/03/2015] [Accepted: 01/04/2015] [Indexed: 01/23/2023] Open
Abstract
Schwann cell (SC) transplantation as a cell-based therapy can enhance peripheral and central nerve repair experimentally, but it is limited by the donor site morbidity for clinical application. We investigated weather respiratory mucosa stem cells (REMSCs), a kind of ectomesenchymal stem cells (EMSCs), isolated from rat nasal septum can differentiate into functional Schwann-like cells (SC-like cells). REMSCs proliferated quickly in vitro and expressed the neural crest markers (nestin, vimentin, SOX10, and CD44). Treated with a mixture of glial growth factors for 7 days, REMSCs differentiated into SC-like cells. The differentiated REMSCs (dREMSCs) exhibited a spindle-like morphology similar to SC cells. Immunocytochemical staining and Western blotting indicated that SC-like cells expressed the glial markers (GFAP, S100β, Galc, and P75) and CNPase. When cocultured with dREMSCs for 5 days, PC12 cells differentiated into mature neuron-like cells with long neurites. More importantly, dREMSCs could form myelin structures with the neurites of PC12 cells at 21 days in vitro. Our data indicated that REMSCs, a kind of EMSCs, could differentiate into SC-like cells and have the ability to promote the differentiation of PC12 cells and form myelin in vitro.
Collapse
|
54
|
Wang H, Liu NK, Zhang YP, Deng L, Lu QB, Shields CB, Walker MJ, Li J, Xu XM. Treadmill training induced lumbar motoneuron dendritic plasticity and behavior recovery in adult rats after a thoracic contusive spinal cord injury. Exp Neurol 2015; 271:368-78. [PMID: 26164199 DOI: 10.1016/j.expneurol.2015.07.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/01/2015] [Accepted: 07/04/2015] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is devastating, causing sensorimotor impairments and paralysis. Persisting functional limitations on physical activity negatively affect overall health in individuals with SCI. Physical training may improve motor function by affecting cellular and molecular responses of motor pathways in the central nervous system (CNS) after SCI. Although motoneurons form the final common path for motor output from the CNS, little is known concerning the effect of exercise training on spared motoneurons below the level of injury. Here we examined the effect of treadmill training on morphological, trophic, and synaptic changes in the lumbar motoneuron pool and on behavior recovery after a moderate contusive SCI inflicted at the 9th thoracic vertebral level (T9) using an Infinite Horizon (IH, 200 kDyne) impactor. We found that treadmill training significantly improved locomotor function, assessed by Basso-Beattie-Bresnahan (BBB) locomotor rating scale, and reduced foot drops, assessed by grid walking performance, as compared with non-training. Additionally, treadmill training significantly increased the total neurite length per lumbar motoneuron innervating the soleus and tibialis anterior muscles of the hindlimbs as compared to non-training. Moreover, treadmill training significantly increased the expression of a neurotrophin brain-derived neurotrophic factor (BDNF) in the lumbar motoneurons as compared to non-training. Finally, treadmill training significantly increased synaptic density, identified by synaptophysin immunoreactivity, in the lumbar motoneuron pool as compared to non-training. However, the density of serotonergic terminals in the same regions did not show a significant difference between treadmill training and non-training. Thus, our study provides a biological basis for exercise training as an effective medical practice to improve recovery after SCI. Such an effect may be mediated by synaptic plasticity, and neurotrophic modification in the spared lumbar motoneuron pool caudal to a thoracic contusive SCI.
Collapse
Affiliation(s)
- Hongxing Wang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, PR China; Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Yi Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY 40202, United States
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Qing-Bo Lu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Christopher B Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY 40202, United States
| | - Melissa J Walker
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jianan Li
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, PR China.
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| |
Collapse
|
55
|
Abstract
ABSTRACT Restoration of lost neuronal function after spinal cord injury still remains a considerable challenge for current medicine. Over the last decade, regenerative medicine has recorded rapid and promising advancements in stem cell research, genetic engineering and the progression of new sophisticated biomaterials as well as nanotechnology. This advancement has also been reflected in neural tissue engineering, where, along with the development of a new generation of well-designed biopolymer scaffolds, multifactorial therapeutic strategies are being validated in order to determine the greatest possible repair efficacy of the complex CNS pathophysiology. Much attention is currently focused on the designing of multifunctional polymer scaffolds as systems for targeted drug or gene delivery, electrical stimulation or as substrates creating a special micro-environment, promoting the growth and desired differentiation of various cell lines. In this review, the latest advances in biomaterial technology together with various combinatorial strategies designed to treat spinal cord injury treatment are summarized and discussed.
Collapse
|
56
|
Mallory GW, Grahn PJ, Hachmann JT, Lujan JL, Lee KH. Optical stimulation for restoration of motor function after spinal cord injury. Mayo Clin Proc 2015; 90:300-7. [PMID: 25659246 PMCID: PMC4339262 DOI: 10.1016/j.mayocp.2014.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 12/31/2022]
Abstract
Spinal cord injury can be defined as a loss of communication between the brain and the body due to disrupted pathways within the spinal cord. Although many promising molecular strategies have emerged to reduce secondary injury and promote axonal regrowth, there is still no effective cure, and recovery of function remains limited. Functional electrical stimulation (FES) represents a strategy developed to restore motor function without the need for regenerating severed spinal pathways. Despite its technological success, however, FES has not been widely integrated into the lives of spinal cord injury survivors. In this review, we briefly discuss the limitations of existing FES technologies. Additionally, we discuss how optogenetics, a rapidly evolving technique used primarily to investigate select neuronal populations within the brain, may eventually be used to replace FES as a form of therapy for functional restoration after spinal cord injury.
Collapse
Affiliation(s)
- Grant W Mallory
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN
| | - Peter J Grahn
- Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Jan T Hachmann
- School of Medicine, Heidelberg University, Neuenheimer Feld, Bergheim, Germany
| | - J Luis Lujan
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Kendall H Lee
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN.
| |
Collapse
|
57
|
Liu J, Zhang SQ, Wu MF, Piao Z, Yao J, Li JH, Wang XG. Edaravone combined with Schwann cell transplantation may repair spinal cord injury in rats. Neural Regen Res 2015; 10:230-6. [PMID: 25883621 PMCID: PMC4392670 DOI: 10.4103/1673-5374.152376] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2014] [Indexed: 11/04/2022] Open
|
58
|
Abstract
Stem cell-based interventions aim to use special regenerative cells (stem cells) to facilitate neuronal function beyond the site of the injury. Many studies involving animal models of spinal cord injury (SCI) suggest that certain stem cell-based therapies may restore function after SCI. Currently, in case of spinal cord injuries, new discoveries with clinical implications have been continuously made in basic stem cell research, and stem cell-based approaches are advancing rapidly toward application in patients. There is a huge base of preclinical evidence in vitro and in animal models which suggests the safety and clinical efficacy of cellular therapies after SCI. Despite this, data from clinical studies is not very encouraging and at times confounding. Here, we have attempted to cover preclinical and clinical evidence base dealing with safety, feasibility and efficacy of cell based interventions after SCI. The limitations of preclinical data and the reasons underlying its failure to translate in a clinical setting are also discussed. Based on the evidence base, it is suggested that a multifactorial approach is required to address this situation. Need for standardized, stringently designed multi-centric clinical trials for obtaining validated proof of evidence is also highlighted.
Collapse
Affiliation(s)
- Harvinder Singh Chhabra
- Spine Service, Indian Spinal Injuries Centre, Vasant Kunj, New Delhi, India,Address for correspondence: Dr. Harvinder Singh Chhabra, Indian Spinal Injuries Centre, Sector C, Vasant Kunj, New Delhi - 110 070, India. E-mail:
| | - Kanchan Sarda
- Spine Service, Indian Spinal Injuries Centre, Vasant Kunj, New Delhi, India
| |
Collapse
|
59
|
Walker CL, Wang X, Bullis C, Liu NK, Lu Q, Fry C, Deng L, Xu XM. Biphasic bisperoxovanadium administration and Schwann cell transplantation for repair after cervical contusive spinal cord injury. Exp Neurol 2014; 264:163-72. [PMID: 25510318 DOI: 10.1016/j.expneurol.2014.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 11/30/2014] [Accepted: 12/03/2014] [Indexed: 11/20/2022]
Abstract
Schwann cells (SCs) hold promise for spinal cord injury (SCI) repair; however, there are limitations for its use as a lone treatment. We showed that acute inhibition of the phosphatase and tensin homolog deleted on chromosome ten (PTEN) by bisperoxovanadium (bpV) was neuroprotective and enhanced function following cervical hemicontusion SCI. We hypothesized that combining acute bpV therapy and delayed SC engraftment would further improve neuroprotection and recovery after cervical SCI. Adult female Sprague-Dawley (SD) rats were randomly sorted into 5 groups: sham, vehicle, bpV, SC transplantation, and bpV+SC transplantation. SCs were isolated from adult green fluorescent protein (GFP)-expressing SD rats (GFP-SCs). 200 μg/kg bpV(pic) was administered intraperitoneally (IP) twice daily for 7 days post-SCI in bpV-treated groups. GFP-SCs (1×10(6) in 5 μl medium) were transplanted into the lesion epicenter at the 8th day post-SCI. Forelimb function was tested for 10 weeks and histology was assessed. bpV alone significantly reduced lesion (by 40%, p<0.05) and cavitation (by 65%, p<0.05) and improved functional recovery (p<0.05) compared to injury alone. The combination promoted similar neuroprotection (p<0.01 vs. injury); however, GFP-SCs alone did not. Both SC-transplanted groups exhibited remarkable long-term SC survival, SMI-31(+) axon ingrowth and RECA-1(+) vasculature presence in the SC graft; however, bpV+SCs promoted an 89% greater axon-to-lesion ratio than SCs only. We concluded that bpV likely contributed largely to the neuroprotective and functional benefits while SCs facilitated considerable host-tissue interaction and modification. The combination of the two shows promise as an attractive strategy to enhance recovery after SCI.
Collapse
Affiliation(s)
- Chandler L Walker
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiaofei Wang
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Carli Bullis
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qingbo Lu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Colin Fry
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
60
|
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
|
61
|
Deng LX, Walker C, Xu XM. Schwann cell transplantation and descending propriospinal regeneration after spinal cord injury. Brain Res 2014; 1619:104-14. [PMID: 25257034 DOI: 10.1016/j.brainres.2014.09.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/08/2014] [Accepted: 09/15/2014] [Indexed: 01/15/2023]
Abstract
After spinal cord injury (SCI), poor ability of damaged axons of the central nervous system (CNS) to regenerate causes very limited functional recovery. Schwann cells (SCs) have been widely explored as promising donors for transplantation to promote axonal regeneration in the CNS including the spinal cord. Compared with other CNS axonal pathways, injured propriospinal tracts display the strongest regenerative response to SC transplantation. Even without providing additional neurotrophic factors, propriospinal axons can grow into the SC environment which is rarely seen in supraspinal tracts. Propriospinal tract has been found to respond to several important neurotrophic factors secreted by SCs. Therefore, the SC is considered to be one of the most promising candidates for cell-based therapies for SCI. Since many reviews have already appeared on topics of SC transplantation in SCI repair, this review will focus particularly on the rationale of SC transplantation in mediating descending propriospinal axonal regeneration as well as optimizing such regeneration by using different combinatorial strategies. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Ling-Xiao Deng
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Chandler Walker
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiao-Ming Xu
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
62
|
Zong H, Zhao H, Zhao Y, Jia J, Yang L, Ma C, Zhang Y, Dong Y. Nanoparticles carrying neurotrophin-3-modified Schwann cells promote repair of sciatic nerve defects. Neural Regen Res 2014; 8:1262-8. [PMID: 25206420 PMCID: PMC4107647 DOI: 10.3969/j.issn.1673-5374.2013.14.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 04/25/2013] [Indexed: 12/03/2022] Open
Abstract
Schwann cells and neurotrophin-3 play an important role in neural regeneration, but the secretion of neurotrophin-3 from Schwann cells is limited, and exogenous neurotrophin-3 is inactived easily in vivo. In this study, we have transfected neurotrophin-3 into Schwann cells cultured in vitro using nanoparticle liposomes. Results showed that neurotrophin-3 was successfully transfected into Schwann cells, where it was expressed effectively and steadily. A composite of Schwann cells transfected with neurotrophin-3 and poly(lactic-co-glycolic acid) biodegradable conduits was transplanted into rats to repair 10-mm sciatic nerve defects. Transplantation of the composite scaffold could restore the myoelectricity and wave amplitude of the sciatic nerve by electrophysiological examination, promote nerve axonal and myelin regeneration, and delay apoptosis of spinal motor neurons. Experimental findings indicate that neurotrophin-3 transfected Schwann cells combined with bridge grafting can promote neural regeneration and functional recovery after nerve injury.
Collapse
Affiliation(s)
- Haibin Zong
- Functional Laboratory, School of Basic Medical Sciences, Xinxiang Medical College, Xinxiang 453003, Henan Province, China
| | - Hongxing Zhao
- Department of Orthopedics, the First Affiliated Hospital of Xinxiang Medical College, Weihui 453100, Henan Province, China
| | - Yilei Zhao
- Department of Orthopedics, the First Affiliated Hospital of Xinxiang Medical College, Weihui 453100, Henan Province, China
| | - Jingling Jia
- Department of Orthopedics, the First Affiliated Hospital of Xinxiang Medical College, Weihui 453100, Henan Province, China
| | - Libin Yang
- Department of Orthopedics, the First Affiliated Hospital of Xinxiang Medical College, Weihui 453100, Henan Province, China
| | - Chao Ma
- Department of Orthopedics, the First Affiliated Hospital of Xinxiang Medical College, Weihui 453100, Henan Province, China
| | - Yang Zhang
- Functional Laboratory, School of Basic Medical Sciences, Xinxiang Medical College, Xinxiang 453003, Henan Province, China
| | - Yuzhen Dong
- Department of Orthopedics, the First Affiliated Hospital of Xinxiang Medical College, Weihui 453100, Henan Province, China
| |
Collapse
|
63
|
Jesuraj NJ, Marquardt LM, Kwasa JA, Sakiyama-Elbert SE. Glial cell line-derived neurotrophic factor promotes increased phenotypic marker expression in femoral sensory and motor-derived Schwann cell cultures. Exp Neurol 2014; 257:10-8. [PMID: 24731946 PMCID: PMC4065822 DOI: 10.1016/j.expneurol.2014.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/06/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
Abstract
Schwann cells (SCs) secrete growth factors and extracellular matrix molecules that promote neuronal survival and help guide axons during regeneration. Transplantation of SCs is a promising strategy for enhancing peripheral nerve regeneration. However, we and others have shown that after long-term in vitro expansion, SCs revert to a de-differentiated state similar to the phenotype observed after injury. In vivo, glial cell-line derived neurotrophic factor (GDNF) may guide the differentiation of SCs to remyelinate regenerating axons. Therefore, we hypothesized that exogenous GDNF may guide the differentiation of SCs into their native phenotypes in vitro through stimulation of GDNF family receptor (GFR)α-1. When activated in SCs, GFRα-1 promotes phosphorylation of Fyn, a Src family tyrosine kinase responsible for mediating downstream signaling for differentiation and proliferation. In this study, SCs harvested from the sensory and motor branches of rat femoral nerve were expanded in vitro and then cultured with 50 or 100ng/mL of GDNF. The exogenous GDNF promoted differentiation of sensory and motor-derived SCs back to their native phenotypes, as demonstrated by decreased proliferation after 7days and increased expression of S100Ββ and phenotype-specific markers. Furthermore, inhibiting Fyn with Src family kinase inhibitors, PP2 and SU6656, and siRNA-mediated knockdown of Fyn reduced GDNF-stimulated differentiation of sensory and motor-derived SCs. These results demonstrate that activating Fyn is necessary for GDNF-stimulated differentiation of femoral nerve-derived SCs into their native phenotypes in vitro. Therefore GDNF could be incorporated into SC-based therapies to promote differentiation of SCs into their native phenotype to improve functional nerve regeneration.
Collapse
Affiliation(s)
- Nithya J Jesuraj
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Laura M Marquardt
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Jasmine A Kwasa
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Shelly E Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
64
|
Wang X, Xu XM. Long-term survival, axonal growth-promotion, and myelination of Schwann cells grafted into contused spinal cord in adult rats. Exp Neurol 2014; 261:308-19. [PMID: 24873728 DOI: 10.1016/j.expneurol.2014.05.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 05/18/2014] [Accepted: 05/20/2014] [Indexed: 01/25/2023]
Abstract
Schwann cells (SCs) have been considered to be one of the most promising cell types for transplantation to treat spinal cord injury (SCI) due to their unique growth-promoting properties. Despite the extensive use as donor cells for transplantation in SCI models, the fate of SCs is controversial due in part to the lack of a reliable marker for tracing the grafted SCs. To precisely assess the fate and temporal profile of transplanted SCs, we isolated purified SCs from sciatic nerves of adult transgenic rats overexpressing GFP (SCs-GFP). SCs-GFP were directly injected into the epicenter of a moderate contusive SCI at the mid-thoracic level at 1week post-injury. The number of SCs-GFP or SCs-GFP labeled with Bromodeoxyuridine (BrdU) was quantified at 5min, 1day, and 1, 2, 4, 12 and 24weeks after cell injection. Basso, Beattie, and Bresnahan (BBB) locomotor rating scale, footfall error, thermal withdrawal latency, and footprint analysis were performed before and after the SCs-GFP transplantation. After transplantation, SCs-GFP quickly filled the lesion cavity. A remarkable survival of grafted SCs-GFP up to 24weeks post-grafting was observed with clearly identified SC individuals. SCs-GFP proliferated after injection, peaked at 2weeks (26% of total SCs-GFP), decreased thereafter, and ceased at 12weeks post-grafting. Although grafted SCs-GFP were mainly confined within the border of surrounding host tissue, they migrated along the central canal for up to 5.0mm at 4weeks post-grafting. Within the lesion site, grafted SCs-GFP myelinated regenerated axons and expressed protein zero (P0) and myelin basic protein (MBP). Within the SCs-GFP grafts, new blood vessels were formed. Except for a significant decrease of angle of rotation in the footprint analysis, we did not observe significant behavioral improvements in BBB locomotor rating scale, thermal withdrawal latency, or footfall errors, compared to the control animals that received no SCs-GFP. We conclude that SCs-GFP can survive remarkably well, proliferate, migrate along the central canal, and myelinate regenerated axons when being grafted into a clinically-relevant contusive SCI in adult rats. Combinatorial strategies, however, are essential to achieve a more meaningful functional regeneration of which SCs may play a significant role.
Collapse
Affiliation(s)
- Xiaofei Wang
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
65
|
Fon D, Al‐Abboodi A, Chan PPY, Zhou K, Crack P, Finkelstein DI, Forsythe JS. Effects of GDNF-loaded injectable gelatin-based hydrogels on endogenous neural progenitor cell migration. Adv Healthc Mater 2014; 3:761-74. [PMID: 24596339 DOI: 10.1002/adhm.201300287] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/11/2013] [Indexed: 12/21/2022]
Abstract
Brain repair following disease and injury is very limited due to difficulties in recruiting and mobilizing stem cells towards the lesion. More importantly, there is a lack of structural and trophic support to maintain viability of the limited stem/progenitor cells present. This study investigates the effectiveness of an injectable gelatin-based hydrogel in attracting neural progenitor cells (NPCs) from the subventricular zone (SVZ) towards the implant. Glial cell-line-derived neurotrophic factor (GDNF) encapsulated within the hydrogel and porosity within the hydrogel prevents glial scar formation. By directly targeting the hydrogel implant towards the SVZ, neuroblasts can actively migrate towards and along the implant tract. Significantly more doublecortin (DCX)-positive neuroblasts surround implants at 7 d post-implantation (dpi) compared with lesion alone controls, an effect that is enhanced when GDNF is incorporated into the hydrogels. Neuroblasts are not observed at the implant boundary at 21 dpi, indicating that neuroblast migration has halted, and neuroblasts have either matured or have not survived. The development of an injectable gelatin-based hydrogel has significant implications for the treatment of some neurodegenerative diseases and brain injuries. The ability of GDNF and porosity to effectively prevent glial scar formation will allow better integration and interaction between the implant and surrounding neural tissue.
Collapse
Affiliation(s)
- Deniece Fon
- Department of Materials Engineering Monash University Clayton VIC 3800 Australia
| | - Aswan Al‐Abboodi
- Department of Chemical Engineering Monash University Clayton VIC 3800 Australia
- Micro/Nanophysics Research Laboratory RMIT University Melbourne VIC 3000 Australia
| | - Peggy P. Y. Chan
- Micro/Nanophysics Research Laboratory RMIT University Melbourne VIC 3000 Australia
- Melbourne Centre for Nanofabrication Clayton VIC 3168 Australia
| | - Kun Zhou
- Department of Materials Engineering Monash University Clayton VIC 3800 Australia
| | - Peter Crack
- Department of Pharmacology The University of Melbourne Parkville VIC 3010 Australia
| | - David I. Finkelstein
- Florey Institute of Neuroscience and Mental Health, Parkville The University of Melbourne VIC 3010 Australia
| | - John S. Forsythe
- Department of Materials Engineering Monash University Clayton VIC 3800 Australia
| |
Collapse
|
66
|
Abstract
Traumatic spinal cord injury (SCI) results in a cascade of tissue responses leading to cell death, axonal degeneration, and glial scar formation, exacerbating the already hostile environment and further inhibiting axon regeneration. Overcoming these inhibitory cues and promoting axonal regeneration is one of the primary targets in developing a cure for SCI. Previously, we demonstrated that transplantation of bone morphogenetic protein (BMP)-induced astrocytes derived from embryonic glial-restricted precursors (GDAs(BMP)) promotes extensive axonal growth and motor function recovery in a rodent spinal cord injury model. Here, we identify periostin (POSTN), a secreted protein, as a key component of GDA(BMP)-induced axonal regeneration. POSTN is highly expressed by GDAs(BMP) and the perturbation of POSTN expression by shRNA diminished GDA(BMP)-induced neurite extension in vitro. We also found that recombinant POSTN is sufficient to overcome the inhibitory effect of scar-associated molecules and promote neurite extension in vitro by signaling through focal adhesion kinase and Akt. Furthermore, transplantation of POSTN-deficient GDAs(BMP) into the injured rat spinal cord resulted in compromised axonal regeneration, indicating that POSTN plays an essential role in GDA(BMP)-mediated axonal regeneration. This finding reveals not only one of the major mechanisms underlying GDA(BMP)-dependent recovery from SCI, but also the potential of POSTN as a therapeutic agent for traumatic injury of the CNS.
Collapse
|
67
|
McKay CA, Pomrenke R, McLane JS, Schaub NJ, DeSimone EK, Ligon LA, Gilbert RJ. An injectable, calcium responsive composite hydrogel for the treatment of acute spinal cord injury. ACS APPLIED MATERIALS & INTERFACES 2014; 6:1424-38. [PMID: 24397537 PMCID: PMC3982972 DOI: 10.1021/am4027423] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 01/03/2014] [Indexed: 05/09/2023]
Abstract
Immediately following spinal cord injury, further injury can occur through several secondary injury cascades. As a consequence of cell lysis, an increase in extracellular Ca(2+) results in additional neuronal loss by inducing apoptosis. Thus, hydrogels that reduce extracellular Ca(2+) concentration may reduce secondary injury severity. The goal of this study was to develop composite hydrogels consisting of alginate, chitosan, and genipin that interact with extracellular Ca(2+) to enable in situ gelation while maintaining an elastic modulus similar to native spinal cord (∼1000 Pa). It was hypothesized that incorporation of genipin and chitosan would regulate hydrogel electrostatic characteristics and influence hydrogel porosity, degradation, and astrocyte behavior. Hydrogel composition was varied to create hydrogels with statistically similar mechanical properties (∼1000 Pa) that demonstrated tunable charge characteristics (6-fold range in free amine concentration) and degradation rate (complete degradation between 7 and 28 days; some blends persist after 28 days). Hydrogels demonstrate high sensitivity to Ca(2+) concentration, as a 1 mM change during fabrication induced a significant change in elastic modulus. Additionally, hydrogels incubated in a Ca(2+)-containing solution exhibited an increased linear viscoelastic limit (LVE) and an increased elastic modulus above the LVE limit in a time dependent manner. An extension of the LVE limit implies a change in hydrogel cross-linking structure. Attachment assays demonstrated that addition of chitosan/genipin to alginate hydrogels induced up to a 4-fold increase in the number of attached astrocytes and facilitated astrocyte clustering on the hydrogel surface in a composition dependent manner. Furthermore, Western blots demonstrated tunable glial fibrillary acid protein (GFAP) expression in astrocytes cultured on hydrogel blends, with some hydrogel compositions demonstrating no significant increase in GFAP expression compared to astrocytes cultured on glass. Thus, alginate/chitosan/genipin hydrogel composites show promise as scaffolds that regulate astrocyte behavior and for the prevention of Ca(2+)-related secondary neuron damage during acute SCI.
Collapse
Affiliation(s)
- Christopher A. McKay
- Center for Biotechnology and Interdisciplinary
Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, 12180-3590 United States
| | - Rebecca
D. Pomrenke
- Center for Biotechnology and Interdisciplinary
Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, 12180-3590 United States
| | - Joshua S. McLane
- Center for Biotechnology and Interdisciplinary
Studies, Department of Biology, Rensselaer
Polytechnic Institute, Troy, New York, 12180-3590 United States
| | - Nicholas J. Schaub
- Center for Biotechnology and Interdisciplinary
Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, 12180-3590 United States
| | - Elise K. DeSimone
- Center for Biotechnology and Interdisciplinary
Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, 12180-3590 United States
| | - Lee A. Ligon
- Center for Biotechnology and Interdisciplinary
Studies, Department of Biology, Rensselaer
Polytechnic Institute, Troy, New York, 12180-3590 United States
| | - Ryan J. Gilbert
- Center for Biotechnology and Interdisciplinary
Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, 12180-3590 United States
| |
Collapse
|
68
|
Lukovic D, Valdés-Sanchez L, Sanchez-Vera I, Moreno-Manzano V, Stojkovic M, Bhattacharya SS, Erceg S. Brief Report: Astrogliosis Promotes Functional Recovery of Completely Transected Spinal Cord Following Transplantation of hESC-Derived Oligodendrocyte and Motoneuron Progenitors. Stem Cells 2014; 32:594-599. [DOI: 10.1002/stem.1562] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Spinal cord injury results in neural loss and consequently motor and sensory impairment below the injury. Reactive astrocytes contribute to formation of glial scar, thus impeding axonal regeneration, through secretion of extracellular matrix molecules, chondroitin sulfate proteoglycans (CSPGs). In this study, we analyze lesion site tissue to reveal the possible mechanism underlying the functional recovery after cell transplantation of human embryonic stem cell (hESC)-derived oligodendrocyte progenitor cell (OPC) and motoneuron progenitors (MP) and propose that transplanted cells increase astrogliosis through the regenerative signaling pathways activated in the host tissue that may crucial for restoring locomotor ability. We show that the transplantation of hESC-derived OPC and MP promotes astrogliosis, through activation of Jagged1-dependent Notch and Jak/STAT signaling that support axonal survival. The transplanted cells in synergism with reactive astrocytes create permissive environment in which the expression of detrimental genes (Cspg, Tenascins, and genes involved in SLIT/ROBO signaling) was significantly decreased while expression of beneficial ones (Laminins and Fibronectin) was increased. According to our data, this mechanism is activated in all transplantation groups independently of the level of locomotor recovery. These results indicate that modifying the beneficial function of reactive astrocytes could be a feasible therapeutic strategy for spinal cord injury in future. Stem Cells 2014;32:594–599
Collapse
Affiliation(s)
- Dunja Lukovic
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | - Lourdes Valdés-Sanchez
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | - Irene Sanchez-Vera
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | | | - Miodrag Stojkovic
- Spebo Medical, Leskovac, Serbia
- Human Genetics Faculty of Medical Sciences, Kragujevac, Serbia
| | - Shomi S. Bhattacharya
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | - Slaven Erceg
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| |
Collapse
|
69
|
Aquaporin-4 mitigates retrograde degeneration of rubrospinal neurons by facilitating edema clearance and glial scar formation after spinal cord injury in mice. Mol Neurobiol 2014; 49:1327-37. [PMID: 24390474 DOI: 10.1007/s12035-013-8607-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/08/2013] [Indexed: 02/02/2023]
Abstract
Atrophy of upper motor neurons hampers axonal regeneration and functional recovery following spinal cord injury (SCI). Apart from the severity of primary injury, a series of secondary pathological damages including spinal cord edema and glial scar formation affect the fate of injured upper motor neurons. The aquaporin-4 (AQP4) water channel plays a critical role in water homeostasis and migration of astrocytes in the central nervous system, probably offering a new therapeutic target for protecting against upper motor neuron degeneration after SCI. To test this hypothesis, we examined the effect of AQP4 deficiency on atrophy of rubrospinal neurons after unilateral rubrospinal tract transection at the fourth cervical level in mice. AQP4 gene knockout (AQP4-/-) mice exhibited high extent of spinal cord edema at 72 h after lesion compared with wild-type littermates. AQP4-/- mice showed impairments in astrocyte migration toward the transected site with a greater lesion volume at 1 week after surgery and glial scar formation with a larger cyst volume at 6 weeks. More severe atrophy and loss of axotomized rubrospinal neurons as well as axonal degeneration in the rubrospinal tract rostral to the lesion were observed in AQP4-/- mice at 6 weeks after SCI. AQP4 expression was downregulated at the lesioned spinal segment at 3 days and 1 week after injury, but upregulated at 6 weeks. These results demonstrated that AQP4 not only mitigates spinal cord damage but also ameliorates retrograde degeneration of rubrospinal neurons by promoting edema clearance and glial scar formation after laceration SCI. This finding supports the notion that AQP4 may be a promising therapeutic target for SCI.
Collapse
|
70
|
Combined treatment with platelet-rich plasma and brain-derived neurotrophic factor-overexpressing bone marrow stromal cells supports axonal remyelination in a rat spinal cord hemi-section model. Cytotherapy 2013; 15:792-804. [PMID: 23731762 DOI: 10.1016/j.jcyt.2013.04.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 03/24/2013] [Accepted: 04/07/2013] [Indexed: 11/21/2022]
Abstract
BACKGROUND AIMS Combining biologic matrices is becoming a better choice to advance stem cell-based therapies. Platelet-rich plasma (PRP) is a biologic product of concentrated platelets and has been used to promote regeneration of peripheral nerves after injury. We examined whether PRP could induce rat bone marrow stromal cells (BMSCs) differentiation in vitro and whether a combination of BMSCs, PRP and brain-derived neurotrophic factor (BDNF) could provide additive therapeutic benefits in vivo after spinal cord injury (SCI). METHODS BMSCs and BDNF-secreting BMSCs (BDNF-BMSCs) were cultured with PRP for 7 days and 21 days, respectively, and neurofilament (NF)-200, glial fibrillary acidic protein (GFAP), microtubule-associated protein 2 (MAP2) and ribosomal protein S6 kinase (p70S6K) gene levels were assessed. After T10 hemi-section in 102 rats, 15-μL scaffolds (PRP alone, BMSCs, PRP/BMSCs, BDNF-BMSCs or PRP/BDNF-BMSCs) were transplanted into the lesion area, and real-time polymerase chain reaction, Western blot, immunohistochemistry and ultrastructural studies were performed. RESULTS The messenger RNA expression of NF-200, GFAP, MAP2 and p70S6K was promoted in BMSCs and BDNF-BMSCs after culture with PRP in vitro. BDNF levels were significantly higher in the injured spinal cord after implantation of BDNF-BMSCs. In the PRP/BDNF-BMSCs group at 8 weeks postoperatively, more GFAP was observed, with less accumulation of astrocytes at the graft-host interface. Rats that received PRP and BDNF-BMSC implants showed enhanced hind limb locomotor performance at 8 weeks postoperatively compared with control animals, with more axonal remyelination. CONCLUSIONS A combined treatment comprising PRP and BDNF-overexpressing BMSCs produced beneficial effects in rats with regard to functional recovery after SCI through enhancing migration of astrocytes into the transplants and axonal remyelination.
Collapse
|
71
|
Combination of Chondroitinase ABC, Glial Cell Line–Derived Neurotrophic Factor and Nogo A Antibody Delayed-Release Microspheres Promotes the Functional Recovery of Spinal Cord Injury. J Craniofac Surg 2013; 24:2153-7. [DOI: 10.1097/01.scs.0000436700.65891.3b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
72
|
Williams RR, Henao M, Pearse DD, Bunge MB. Permissive Schwann cell graft/spinal cord interfaces for axon regeneration. Cell Transplant 2013; 24:115-31. [PMID: 24152553 DOI: 10.3727/096368913x674657] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The transplantation of autologous Schwann cells (SCs) to repair the injured spinal cord is currently being evaluated in a clinical trial. In support, this study determined properties of spinal cord/SC bridge interfaces that enabled regenerated brainstem axons to cross them, possibly leading to improvement in rat hindlimb movement. Fluid bridges of SCs and Matrigel were placed in complete spinal cord transections. Compared to pregelled bridges of SCs and Matrigel, they improved regeneration of brainstem axons across the rostral interface. The regenerating brainstem axons formed synaptophysin(+) bouton-like terminals and contacted MAP2A(+) dendrites at the caudal interface. Brainstem axon regeneration was directly associated with glial fibrillary acidic protein (GFAP(+)) astrocyte processes that elongated into the SC bridge. Electron microscopy revealed that axons, SCs, and astrocytes were enclosed together within tunnels bounded by a continuous basal lamina. Neuroglycan (NG2) expression was associated with these tunnels. One week after injury, the GFAP(+) processes coexpressed nestin and brain lipid-binding protein, and the tips of GFAP(+)/NG2(+) processes extended into the bridges together with the regenerating brainstem axons. Both brainstem axon regeneration and number of GFAP(+) processes in the bridges correlated with improvement in hindlimb locomotion. Following SCI, astrocytes may enter a reactive state that prohibits axon regeneration. Elongation of astrocyte processes into SC bridges, however, and formation of NG2(+) tunnels enable brainstem axon regeneration and improvement in function. It is important for spinal cord repair to define conditions that favor elongation of astrocytes into lesions/transplants.
Collapse
Affiliation(s)
- Ryan R Williams
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | | |
Collapse
|
73
|
Nerve regeneration restores supraspinal control of bladder function after complete spinal cord injury. J Neurosci 2013; 33:10591-606. [PMID: 23804083 DOI: 10.1523/jneurosci.1116-12.2013] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
A life-threatening disability after complete spinal cord injury is urinary dysfunction, which is attributable to lack of regeneration of supraspinal pathways that control the bladder. Although numerous strategies have been proposed that can promote the regrowth of severed axons in the adult CNS, at present, the approaches by which this can be accomplished after complete cord transection are quite limited. In the present study, we modified a classic peripheral nerve grafting technique with the use of chondroitinase to facilitate the regeneration of axons across and beyond an extensive thoracic spinal cord transection lesion in adult rats. The novel combination treatment allows for remarkably lengthy regeneration of certain subtypes of brainstem and propriospinal axons across the injury site and is followed by markedly improved urinary function. Our studies provide evidence that an enhanced nerve grafting strategy represents a potential regenerative treatment after severe spinal cord injury.
Collapse
|
74
|
Kuric E, Wieloch T, Ruscher K. Dopamine receptor activation increases glial cell line-derived neurotrophic factor in experimental stroke. Exp Neurol 2013; 247:202-8. [DOI: 10.1016/j.expneurol.2013.04.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/26/2013] [Accepted: 04/30/2013] [Indexed: 12/20/2022]
|
75
|
Ansorena E, De Berdt P, Ucakar B, Simón-Yarza T, Jacobs D, Schakman O, Jankovski A, Deumens R, Blanco-Prieto MJ, Préat V, des Rieux A. Injectable alginate hydrogel loaded with GDNF promotes functional recovery in a hemisection model of spinal cord injury. Int J Pharm 2013; 455:148-58. [PMID: 23916821 DOI: 10.1016/j.ijpharm.2013.07.045] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/15/2013] [Accepted: 07/17/2013] [Indexed: 11/29/2022]
Abstract
We hypothesized that local delivery of GDNF in spinal cord lesion via an injectable alginate hydrogel gelifying in situ would support spinal cord plasticity and functional recovery. The GDNF release from the hydrogel was slowed by GDNF encapsulation in microspheres compared to non-formulated GDNF (free GDNF). When injected in a rat spinal cord hemisection model, more neurofilaments were observed in the lesion when the rats were treated with free GDNF-loaded hydrogels. More growing neurites were detected in the tissues surrounding the lesion when the animals were treated with GDNF microsphere-loaded hydrogels. Intense GFAP (astrocytes), low βIII tubulin (neural cells) and RECA-1 (endothelial cells) stainings were observed for non-treated lesions while GDNF-treated spinal cords presented less GFAP staining and more endothelial and nerve fiber infiltration in the lesion site. The animals treated with free GDNF-loaded hydrogel presented superior functional recovery compared with the animals treated with the GDNF microsphere-loaded hydrogels and non-treated animals.
Collapse
Affiliation(s)
- Eduardo Ansorena
- Université Catholique de Louvain, Louvain Drug Research Institute, Pharmaceutics and Drug delivery Unit, 1200 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Chen CH, Huang SY, Chen NF, Feng CW, Hung HC, Sung CS, Jean YH, Wen ZH, Chen WF. Intrathecal granulocyte colony-stimulating factor modulate glial cell line-derived neurotrophic factor and vascular endothelial growth factor A expression in glial cells after experimental spinal cord ischemia. Neuroscience 2013; 242:39-52. [DOI: 10.1016/j.neuroscience.2013.02.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 01/30/2013] [Accepted: 02/09/2013] [Indexed: 12/20/2022]
|
77
|
A novel growth-promoting pathway formed by GDNF-overexpressing Schwann cells promotes propriospinal axonal regeneration, synapse formation, and partial recovery of function after spinal cord injury. J Neurosci 2013; 33:5655-67. [PMID: 23536080 DOI: 10.1523/jneurosci.2973-12.2013] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Descending propriospinal neurons (DPSN) are known to establish functional relays for supraspinal signals, and they display a greater growth response after injury than do the long projecting axons. However, their regenerative response is still deficient due to their failure to depart from growth supportive cellular transplants back into the host spinal cord, which contains numerous impediments to axon growth. Here we report the construction of a continuous growth-promoting pathway in adult rats, formed by grafted Schwann cells overexpressing glial cell line-derived neurotrophic factor (GDNF). We demonstrate that such a growth-promoting pathway, extending from the axonal cut ends to the site of innervation in the distal spinal cord, promoted regeneration of DPSN axons through and beyond the lesion gap of a spinal cord hemisection. Within the distal host spinal cord, regenerated DPSN axons formed synapses with host neurons leading to the restoration of action potentials and partial recovery of function.
Collapse
|
78
|
Awad BI, Carmody MA, Steinmetz MP. Potential role of growth factors in the management of spinal cord injury. World Neurosurg 2013; 83:120-31. [PMID: 23334003 DOI: 10.1016/j.wneu.2013.01.042] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 01/06/2013] [Accepted: 01/11/2013] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To review central nervous system growth factors and their therapeutic potential and clinical translation into spinal cord injury (SCI), as well as the challenges that have been encountered during clinical development. METHODS A systemic review of the available current and historical literature regarding central nervous system growth factors and clinical trials regarding their use in spinal cord injury was conducted. RESULTS The effectiveness of administering growth factors as a potential therapeutic strategy for SCI has been tested with the use of brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, neurotrophin 3, and neurotrophin-4/5. Delivery of growth factors to injured SC has been tested by numerous methods. Unfortunately, most of clinical trials at this time are uncontrolled and have questionable results because of lack of efficacy and/or unacceptable side effects. CONCLUSIONS There is promise in the use of specific growth factors therapeutically for SCI. However, more studies involving neuronal regeneration and functional recovery are needed, as well the development of delivery methods that allow sufficient quantity of growth factors while restricting their distribution to target sites.
Collapse
Affiliation(s)
- Basem I Awad
- Department of Neurosurgery, Mansoura University School of Medicine, Mansoura, Egypt; Department of Neurosciences, MetroHealth Medical Center, Cleveland, Ohio, USA
| | - Margaret A Carmody
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Michael P Steinmetz
- Department of Neurosciences, MetroHealth Medical Center, Cleveland, Ohio, USA.
| |
Collapse
|
79
|
Li J, Lepski G. Cell transplantation for spinal cord injury: a systematic review. BIOMED RESEARCH INTERNATIONAL 2013; 2013:786475. [PMID: 23484157 PMCID: PMC3581246 DOI: 10.1155/2013/786475] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 11/16/2012] [Accepted: 12/11/2012] [Indexed: 02/07/2023]
Abstract
Cell transplantation, as a therapeutic intervention for spinal cord injury (SCI), has been extensively studied by researchers in recent years. A number of different kinds of stem cells, neural progenitors, and glial cells have been tested in basic research, and most have been excluded from clinical studies because of a variety of reasons, including safety and efficacy. The signaling pathways, protein interactions, cellular behavior, and the differentiated fates of experimental cells have been studied in vitro in detail. Furthermore, the survival, proliferation, differentiation, and effects on promoting functional recovery of transplanted cells have also been examined in different animal SCI models. However, despite significant progress, a "bench to bedside" gap still exists. In this paper, we comprehensively cover publications in the field from the last years. The most commonly utilized cell lineages were covered in this paper and specific areas covered include survival of grafted cells, axonal regeneration and remyelination, sensory and motor functional recovery, and electrophysiological improvements. Finally we also review the literature on the in vivo tracking techniques for transplanted cells.
Collapse
Affiliation(s)
- Jun Li
- Department of Neurosurgery, Eberhard Karls University, 72076 Tübingen, Germany
- Department of Spine Surgery, The Affiliated Hospital of Luzhou Medical College, 646000 Luzhou, China
| | - Guilherme Lepski
- Department of Neurosurgery, Eberhard Karls University, 72076 Tübingen, Germany
- Division of Neurosurgery, Department of Neurology, Faculdade de Medicina, Universidade de São Paulo, Avnida Dr. Enéas de Carvalho Aguiar 255, 05403-000 São Paulo, SP, Brazil
| |
Collapse
|
80
|
Fan C, Zheng Y, Cheng X, Qi X, Bu P, Luo X, Kim DH, Cao Q. Transplantation of D15A-expressing glial-restricted-precursor-derived astrocytes improves anatomical and locomotor recovery after spinal cord injury. Int J Biol Sci 2012; 9:78-93. [PMID: 23289019 PMCID: PMC3535536 DOI: 10.7150/ijbs.5626] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/19/2012] [Indexed: 12/23/2022] Open
Abstract
The transplantation of neural stem/progenitor cells is a promising therapeutic strategy for spinal cord injury (SCI). In this study, we tested whether combination of neurotrophic factors and transplantation of glial-restricted precursor (GRPs)-derived astrocytes (GDAs) could decrease the injury and promote functional recovery after SCI. We developed a protocol to quickly produce a sufficiently large, homogenous population of young astrocytes from GRPs, the earliest arising progenitor cell population restricted to the generation of glia. GDAs expressed the axonal regeneration promoting substrates, laminin and fibronectin, but not the inhibitory chondroitin sulfate proteoglycans (CSPGs). Importantly, GDAs or its conditioned medium promoted the neurite outgrowth of dorsal root ganglion neurons in vitro. GDAs were infected with retroviruses expressing EGFP or multi-neurotrophin D15A and transplanted into the contused adult thoracic spinal cord at 8 days post-injury. Eight weeks after transplantation, the grafted GDAs survived and integrated into the injured spinal cord. Grafted GDAs expressed GFAP, suggesting they remained astrocyte lineage in the injured spinal cord. But it did not express CSPG. Robust axonal regeneration along the grafted GDAs was observed. Furthermore, transplantation of D15A-GDAs significantly increased the spared white matter and decreased the injury size compared to other control groups. More importantly, transplantation of D15A-GDAs significantly improved the locomotion function recovery shown by BBB locomotion scores and Tredscan footprint analyses. However, this combinatorial strategy did not enhance the aberrant synaptic connectivity of pain afferents, nor did it exacerbate posttraumatic neuropathic pain. These results demonstrate that transplantation of D15A-expressing GDAs promotes anatomical and locomotion recovery after SCI, suggesting it may be an effective therapeutic approach for SCI.
Collapse
Affiliation(s)
- Chunling Fan
- Department of Anatomy and Neurobiology, Central South University Xianya Medical School, Changsha, Hunan 410011, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Promoting engraftment of transplanted neural stem cells/progenitors using biofunctionalised electrospun scaffolds. Biomaterials 2012; 33:9188-97. [PMID: 23022345 DOI: 10.1016/j.biomaterials.2012.09.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 09/09/2012] [Indexed: 02/02/2023]
Abstract
With the brain's limited capacity for repair, new and innovative approaches are required to promote regeneration. While neural transplantation for a number of neural disease/injuries have been demonstrated, major limitations in the field include poor cell survival and integration. This, in part, is due to the non-conducive environment of the adult brain, failing to provide adequate chemical and physical support for new neurons. Here we examine the capacity of fibrous poly ε-caprolactone (PCL) scaffolds, biofunctionalised with immobilised glial cell-derived neurotrophic factor (GDNF), to influence primary cortical neural stem cells/progenitors in vitro and enhance integration of these cells following transplantation into the brain parenchyma. Immobilisation of GDNF was confirmed prior to in vitro culturing and at 28 days after implantation into the brain, demonstrating long-term delivery of the protein. In vitro, we demonstrate that PCL with immobilised GDNF (iGDNF) significantly enhances cell viability and neural stem cell/progenitor proliferation compared to conventional 2-dimensional cultureware. Upon implantation, PCL scaffolds including iGDNF enhanced the survival, proliferation, migration, and neurite growth of transplanted cortical cells, whilst suppressing inflammatory reactive astroglia.
Collapse
|
82
|
Theophylline regulates inflammatory and neurotrophic factor signals in functional recovery after C2-hemisection in adult rats. Exp Neurol 2012; 238:79-88. [PMID: 22981449 DOI: 10.1016/j.expneurol.2012.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 08/09/2012] [Accepted: 08/11/2012] [Indexed: 11/24/2022]
Abstract
Recovery of respiratory activity in an upper cervical hemisection model (C2H) of spinal cord injury (SCI) can be induced by systemic theophylline administration 24-48 h after injury. The objectives in the present study are (1) to identify pro-inflammatory and neurotrophic factors expressed after C2H and (2) molecular signals involved in functional recovery. Four groups of adult female rats classified as (i) sham (SH) controls, (ii) subjected to a left C2 hemisection (C2H) only, (iii) C2H rats administered theophylline for 3 consecutive days 2 days after C2H (C2H-T day 5) and (iv) C2H rats treated with theophylline for 3 consecutive days 2 days after C2H and then weaned for 12 days (C2H-T day 17) prior to assessment of respiratory function and molecular analysis were employed. Corresponding sham controls, C2H untreated (vehicle only controls) and C2H treated (theophylline) rats were sacrificed, C3-C6 spinal cord segments quickly dissected and left (ipsilateral) hemi spinal cord and right (contralateral) hemi spinal cord were separately harvested 2 days post surgery. Sham operated and C2H untreated-controls corresponding to C2H-T day 5 and C2H-T day 17 rats, respectively, were prepared similarly. Messenger RNA levels for pro-inflammatory genes (TXNIP, IL-1β, TNF-α and iNOS) and neurotrophic and survival factors (BDNF, GDNF, and Bcl2) were analyzed by real time quantitative PCR. Gene expression pattern was unaltered in SH rats. TXNIP, iNOS, BDNF, GDNF and Bcl2 mRNA levels were significantly increased in the ipsilateral hemi spinal cord in C2H rats. BDNF, GDNF and Bcl2 levels remained elevated in the ipsilateral hemi spinal cord in C2H-T day 5 rats. In this same group, there was further enhancement in TXNIP and IL-1β while iNOS returned to basal levels. Theophylline increased DNA binding activity of transcription factors - cyclic AMP responsive element (CRE) binding protein (CREB) and pro-inflammatory NF-κB. Messenger RNA levels for all genes returned to basal levels in C2H-T day 17 rats. However, BDNF mRNA levels remained significantly elevated after weaning from the drug. Our results suggest that enhanced resolution of early inflammatory processes and expression of pro-survival factors may underlie theophylline-induced respiratory recovery. The results identify potential targets for gene and drug therapies.
Collapse
|
83
|
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
|
84
|
Lin CY, Lee YS, Lin VW, Silver J. Fibronectin inhibits chronic pain development after spinal cord injury. J Neurotrauma 2012; 29:589-99. [PMID: 22022865 DOI: 10.1089/neu.2011.2059] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Chronic pain following spinal cord injury (SCI) is a highly prevalent clinical condition that is difficult to treat. Using both von Frey filaments and radiant infrared heat to assess mechanical allodynia and thermal hyperalgesia, respectively, we have demonstrated that a one-time injection of fibronectin (50 μg/mL) into the spinal dorsal column (1 μL/min each injection for a total of 5 μL) immediately after SCI inhibits the development of mechanical allodynia (but not thermal hyperalgesia) over an 8-month observation period following spinal cord dorsal column crush (DCC). DCC will only induce mechanical Allodynia, but not thermal hyperalgesia or overt motor deficits. By applying various fibronectin fragments as well as competitive inhibitors, these effects were shown to be dependent on the connecting segment-1 (CS-1) motif of fibronectin. Furthermore, we found that acute fibronectin treatment diminished inflammation and blood-spinal cord barrier permeability, which in turn leads to enhanced fiber sparing and sprouting. In particular, the reduction of serotonin (5-HT) in the superficial dorsal horn, an important descending brainstem system in the modulation of pain, was blocked with fibronectin treatment. We conclude that treatment of SCI with fibronectin preserves sensory regulation and prevents the development of chronic allodynia, providing a potential therapeutic intervention to treat chronic pain following SCI.
Collapse
Affiliation(s)
- Ching-Yi Lin
- Department of Neuroscience, Lerner Research Institute, Cleveland, Ohio 44195, USA.
| | | | | | | |
Collapse
|
85
|
|
86
|
Transforming growth factor α transforms astrocytes to a growth-supportive phenotype after spinal cord injury. J Neurosci 2011; 31:15173-87. [PMID: 22016551 DOI: 10.1523/jneurosci.3441-11.2011] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Astrocytes are both detrimental and beneficial for repair and recovery after spinal cord injury (SCI). These dynamic cells are primary contributors to the growth-inhibitory glial scar, yet they are also neuroprotective and can form growth-supportive bridges on which axons traverse. We have shown that intrathecal administration of transforming growth factor α (TGFα) to the contused mouse spinal cord can enhance astrocyte infiltration and axonal growth within the injury site, but the mechanisms of these effects are not well understood. The present studies demonstrate that the epidermal growth factor receptor (EGFR) is upregulated primarily by astrocytes and glial progenitors early after SCI. TGFα directly activates the EGFR on these cells in vitro, inducing their proliferation, migration, and transformation to a phenotype that supports robust neurite outgrowth. Overexpression of TGFα in vivo by intraparenchymal adeno-associated virus injection adjacent to the injury site enhances cell proliferation, alters astrocyte distribution, and facilitates increased axonal penetration at the rostral lesion border. To determine whether endogenous EGFR activation is required after injury, SCI was also performed on Velvet (C57BL/6J-Egfr(Vel)/J) mice, a mutant strain with defective EGFR activity. The affected mice exhibited malformed glial borders, larger lesions, and impaired recovery of function, indicating that intrinsic EGFR activation is necessary for neuroprotection and normal glial scar formation after SCI. By further stimulating precursor proliferation and modifying glial activation to promote a growth-permissive environment, controlled stimulation of EGFR at the lesion border may be considered in the context of future strategies to enhance endogenous cellular repair after injury.
Collapse
|
87
|
Pendleton C, Ahmed I, Quinones-Hinojosa A. Neurotransplantation: lux et veritas, fiction or reality? J Neurosurg Sci 2011; 55:297-304. [PMID: 22198582 PMCID: PMC4659364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Neurotransplantation remains a much-debated frontier in contemporary neurosurgery and neuroscience, with roots dating to the late 19th century. Contemporary applications are far-reaching, and ongoing laboratory research and clinical trials seek to define the mechanisms at play in neurotransplant engraftment and growth, while advancing the field forward into the 21st century. Neural transplantation therapy remains an attractive idea for treating central nervous system (CNS) and peripheral nervous system (PNS) pathologies. Phase I and phase II clinical trials assessing safety and efficacy are currently underway for various disorders. The remainder of this review will focus on ongoing clinical trials and more recent research advances involving neural transplantation therapy for neuronal death, axonal injury, peripheral nerve lesions, and cancer. The field of neural transplantation, while promising, is not without ethical and scientific dilemmas; this review will conclude with a discussion of the challenges researchers and clinicians face as the field of neural transplantation moves forward.
Collapse
Affiliation(s)
- C Pendleton
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, USA
| | | | | |
Collapse
|