201
|
Zhou HL, Yang HJ, Li YM, Wang Y, Yan L, Guo XL, Ba YC, Liu S, Wang TH. Changes in Glial cell line-derived neurotrophic factor expression in the rostral and caudal stumps of the transected adult rat spinal cord. Neurochem Res 2008; 33:927-37. [PMID: 18095158 PMCID: PMC2270371 DOI: 10.1007/s11064-007-9536-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Accepted: 10/23/2007] [Indexed: 02/05/2023]
Abstract
Limited information is available regarding the role of endogenous Glial cell line-derived neurotrophic factor (GDNF) in the spinal cord following transection injury. The present study investigated the possible role of GDNF in injured spinal cords following transection injury (T(9)-T(10)) in adult rats. The locomotor function recovery of animals by the BBB (Basso, Beattie, Bresnahan) scale score showed that hindlimb support and stepping function increased gradually from 7 days post operation (dpo) to 21 dpo. However, the locomotion function in the hindlimbs decreased effectively in GDNF-antibody treated rats. GDNF immunoreactivty in neurons in the ventral horn of the rostral stump was stained strongly at 3 and 7 dpo, and in the caudal stump at 14 dpo, while immunostaining in astrocytes was also seen at all time-points after transection injury. Western blot showed that the level of GDNF protein underwent a rapid decrease at 7 dpo in both stumps, and was followed by a partial recovery at a later time-point, when compared with the sham-operated group. GDNF mRNA-positive signals were detected in neurons of the ventral horn, especially in lamina IX. No regenerative fibers from corticospinal tract can be seen in the caudal segment near the injury site using BDA tracing technique. No somatosensory evoked potentials (SEP) could be recorded throughout the experimental period as well. These findings suggested that intrinsic GDNF in the spinal cord could play an essential role in neuroplasticity. The mechanism may be that GDNF is involved in the regulation of local circuitry in transected spinal cords of adult rats.
Collapse
Affiliation(s)
- Hao-Li Zhou
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041 China
- Institute of Neuroscience, Kunming Medical College, Kunming, 650031 China
| | - Hui-Juan Yang
- Institute of Neuroscience, Kunming Medical College, Kunming, 650031 China
| | - Yong-Mei Li
- Institute of Neuroscience, Kunming Medical College, Kunming, 650031 China
| | - Ying Wang
- Nursing Department, Weifang Medical College, Weifang, 261042 China
| | - Ling Yan
- Institute of Neuroscience, Kunming Medical College, Kunming, 650031 China
| | - Xi-Liang Guo
- Institute of Neuroscience, Kunming Medical College, Kunming, 650031 China
| | - Ying-Chun Ba
- Institute of Neuroscience, Kunming Medical College, Kunming, 650031 China
| | - Su Liu
- Institute of Neuroscience, Kunming Medical College, Kunming, 650031 China
| | - Ting-Hua Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041 China
- Institute of Neuroscience, Kunming Medical College, Kunming, 650031 China
- Department of Histology, Embryology and Neurobiology, College of Preclinical and Forensic Medicine, Sichuan University, Chengdu, 610041 China
| |
Collapse
|
202
|
Deumens R, Joosten EAJ, Waxman SG, Hains BC. Locomotor dysfunction and pain: the scylla and charybdis of fiber sprouting after spinal cord injury. Mol Neurobiol 2008; 37:52-63. [PMID: 18415034 DOI: 10.1007/s12035-008-8016-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Accepted: 03/19/2008] [Indexed: 10/22/2022]
Abstract
Injury to the spinal cord (SCI) can produce a constellation of problems including chronic pain, autonomic dysreflexia, and motor dysfunction. Neuroplasticity in the form of fiber sprouting or the lack thereof is an important phenomenon that can contribute to the deleterious effects of SCI. Aberrant sprouting of primary afferent fibers and synaptogenesis within incorrect dorsal horn laminae leads to the development and maintenance of chronic pain as well as autonomic dysreflexia. At the same time, interruption of connections between supraspinal motor control centers and spinal cord output cells, due to lack of successful regenerative sprouting of injured descending fiber tracts, contributes to motor deficits. Similarities in the molecular control of axonal growth of motor and sensory fibers have made the development of cogent therapies difficult. In this study, we discuss recent findings related to the degradation of inhibitory barriers and promotion of sprouting of motor fibers as a strategy for the restoration of motor function and note that this may induce primary afferent fiber sprouting that can contribute to chronic pain. We highlight the importance of careful attentiveness to off-target molecular- and circuit-level modulation of nociceptive processing while moving forward with the development of therapies that will restore motor function after SCI.
Collapse
Affiliation(s)
- Ronald Deumens
- Pain Management and Research Center, Department of Anesthesiology, Maastricht University Hospital, P. Debyelaan 25, P.O. Box 5800, 6200 AZ, Maastricht, The Netherlands
| | | | | | | |
Collapse
|
203
|
Ritz MF, Hausmann ON. Effect of 17β-estradiol on functional outcome, release of cytokines, astrocyte reactivity and inflammatory spreading after spinal cord injury in male rats. Brain Res 2008; 1203:177-88. [DOI: 10.1016/j.brainres.2008.01.091] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 01/28/2008] [Accepted: 01/29/2008] [Indexed: 11/29/2022]
|
204
|
Itoh T, Satou T, Nishida S, Hashimoto S, Ito H. Immature and mature neurons coexist among glial scars after rat traumatic brain injury. Neurol Res 2008; 29:734-42. [PMID: 18183647 DOI: 10.1179/016164107x208086] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Glial scars around a damaged area after brain injury inhibit neurite elongation from surviving neurons and axonal plasticity, and thus prevent neural network regeneration. However, the generation, differentiation and maturation of neural stem cells (NSCs) among glial scars after brain injury have not yet been reported. METHODS In the present study, we investigated the chronological relationship between gliosis and maturation of new neurons around a damaged area using a rat traumatic brain injury (TBI) model. RESULTS Between 1 and 7 days after injury, many nestin-positive cells were observed around the damaged area. Three days after injury, many small nestin-positive cells showed an astrocytic morphology. Between 1 and 30 days after injury, doublecortin (DCX)-positive cells were present around the damaged area. Three and 7 days after injury, a small number of nestin-positive cells were immunopositive for glial fibrillary acidic protein (GFAP). Seven days after injury, there were DCX-positive cells in the gliosis occurring in the lesion. Thirty days after injury, DCX-positive cells were observed near and among the glial scars and a small number of these cells were immunopositive for NeuN. DISCUSSION These results suggest that DCX-positive cells were present near and among the glial scars after brain injury, and that these cells changed from immature to mature neurons. It is considered that promotion of the maturation and differentiation of newly formed immature neurons near and among glial scars after injury may improve the brain dysfunction induced by glial scars after brain injury.
Collapse
Affiliation(s)
- Tatsuki Itoh
- Department of Pathology, Kinki University School of Medicine, Osaka, Japan.
| | | | | | | | | |
Collapse
|
205
|
Tom VJ, Houlé JD. Intraspinal microinjection of chondroitinase ABC following injury promotes axonal regeneration out of a peripheral nerve graft bridge. Exp Neurol 2008; 211:315-9. [PMID: 18353313 DOI: 10.1016/j.expneurol.2008.01.021] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 12/21/2007] [Accepted: 01/14/2008] [Indexed: 11/19/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPG) within the glial scar formed after central nervous system (CNS) injury are thought to play a crucial role in regenerative failure. We previously showed that delivery of the CSPG-digesting enzyme chondroitinase ABC (ChABC) via an osmotic minipump allowed axonal regeneration and functional recovery in a peripheral nerve graft (PNG)-bridging model. In this study, we sought to overcome the technical limitations associated with minipumps by microinjecting ChABC directly into the distal lesion site in the PN bridging model. Microinjection of ChABC immediately rostral and caudal to an injury site resulted in extensive CSPG digestion. We also demonstrate that this delivery technique is relatively atraumatic and does not result in a noticeable inflammatory response. Importantly, microinjections of ChABC into the lesion site permitted more regenerating axons to exit a PNG and reenter spinal cord tissue than saline injections. These results are similar to our previous findings when ChABC was delivered via a minipump and suggest that microinjecting ChABC is an effective method of delivering the potentially therapeutic enzyme directly to an injury site.
Collapse
Affiliation(s)
- Veronica J Tom
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | | |
Collapse
|
206
|
Kaas JH, Qi HX, Burish MJ, Gharbawie OA, Onifer SM, Massey JM. Cortical and subcortical plasticity in the brains of humans, primates, and rats after damage to sensory afferents in the dorsal columns of the spinal cord. Exp Neurol 2008; 209:407-16. [PMID: 17692844 PMCID: PMC2268113 DOI: 10.1016/j.expneurol.2007.06.014] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Accepted: 06/11/2007] [Indexed: 11/20/2022]
Abstract
The failure of injured axons to regenerate following spinal cord injury deprives brain neurons of their normal sources of activation. These injuries also result in the reorganization of affected areas of the central nervous system that is thought to drive both the ensuing recovery of function and the formation of maladaptive neuronal circuitry. Better understanding of the physiological consequences of novel synaptic connections produced by injury and the mechanisms that control their formation are important to the development of new successful strategies for the treatment of patients with spinal cord injuries. Here we discuss the anatomical, physiological and behavioral changes that take place in response to injury-induced plasticity after damage to the dorsal column pathway in rats and monkeys. Complete section of the dorsal columns of the spinal cord at a high cervical level in monkeys and rats interrupts the ascending axon branches of low threshold mechanoreceptor afferents subserving the forelimb and the rest of the lower body. Such lesions render the corresponding part of the somatotopic representation of primary somatosensory cortex totally unresponsive to tactile stimuli. There are also behavioral consequences of the sensory loss, including an impaired use of the hand/forelimb in manipulating small objects. In monkeys, if some of the afferents from the hand remain intact after dorsal column lesions, these remaining afferents extensively reactivate portions of somatosensory cortex formerly representing the hand. This functional reorganization develops over a postoperative period of 1 month, during which hand use rapidly improves. These recoveries appear to be mediated, at least in part, by the sprouting of preserved afferents within the cuneate nucleus of the dorsal column-trigeminal complex. In rats, such functional collateral sprouting has been promoted by the post-lesion digestion of the perineuronal net in the cuneate nucleus. Thus, this and other therapeutic strategies have the potential of enhancing sensorimotor recoveries after spinal cord injuries in humans.
Collapse
Affiliation(s)
- Jon H Kaas
- Department of Psychology, Vanderbilt University, Nashville, TN 37203, USA.
| | | | | | | | | | | |
Collapse
|
207
|
Rat focal cerebral ischemia induced astrocyte proliferation and delayed neuronal death are attenuated by cyclin-dependent kinase inhibition. J Clin Neurosci 2008; 15:278-85. [PMID: 18207409 DOI: 10.1016/j.jocn.2007.02.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Revised: 01/22/2007] [Accepted: 02/08/2007] [Indexed: 11/23/2022]
Abstract
Astroglial proliferation and delayed neuronal death are two common pathological processes in the ischemic brain. However, it is not clear if astrogliosis causes delayed neuronal death. In this study, we addressed this potential linkage by examining the relationship between attenuated astrocyte proliferation, induced by cyclin-dependent kinase (CDK) inhibition, and delayed neuronal death in rat ischemic hippocampus. Our results show that following middle cerebral artery occlusion (MCAO), astrocyte hypertrophy and proliferation were closely associated with delayed neuronal death. Importantly, administration of olomoucine, a selective CDK inhibitor, not only suppressed astroglial proliferation and glial scar formation, but also decreased neuronal cell death in the ischemic boundary zone and hippocampal CA1 region at days 1 and 30 after MCAO. These results indicate that reactive astrogliosis and delayed neuronal death, at least in rat hippocampus, are sequential pathological events following MCAO. Therefore, suppressing astroglial cell cycle progression in acute focal cerebral ischemia may be beneficial to neuronal survival. Our study also implies that cell cycle regulation should be considered as a promising future therapeutic intervention in treating those neurological diseases characterized by an excessive astrocyte proliferation.
Collapse
|
208
|
NOLIN WESTLEYB, EMMETSBERGER JAIME, BUKHARI NOREEN, ZHANG YAN, LEVINE JOELM, TSIRKA STELLAE. tPA-mediated generation of plasmin is catalyzed by the proteoglycan NG2. Glia 2008; 56:177-89. [PMID: 18000864 PMCID: PMC3895479 DOI: 10.1002/glia.20603] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Paralysis resulting from spinal cord injury is devastating and persistent. One major reason for the inability of the body to heal this type of injury ensues from the local increase of glial cells leading to the formation of a glial scar, and the upregulation of chondroitin sulfate proteoglycans (CSPGs) at the site of injury through which axons are unable to regenerate. Experimental approaches to overcome this problem have accordingly focused on reducing the inhibitory properties of CSPGs, for example by using chondroitinase to remove the sugar chains and reduce the CSPGs to their core protein constituents, although this step alone does not provide dramatic benefits as a monotherapy. Using in vitro and in vivo approaches, we describe here a potentially synergistic therapeutic opportunity based on tissue plasminogen activator (tPA), an extracellular protease that converts plasminogen (plg) into the active protease plasmin. We show that tPA and plg both bind to the CSPG protein NG2, which functions as a scaffold to accelerate the tPA-driven conversion of plg to plasmin. The binding occurs via the tPA and plg kringle domains to domain 2 of the NG2 CSPG core protein, and is enhanced in some settings after chondroitinase-mediated removal of the NG2 proteoglycan side chains. Once generated, plasmin then degrades NG2, both in an in vitro setting using recombinant protein, and in vivo models of spinal cord injury. Our finding that the tPA and plg binding is in some instances more efficient after exposure of the NG2 proteoglycan to chondroitinase treatment suggests that a combined therapeutic approach employing both chondroitinase and the tPA/plasmin proteolytic system could be of significant benefit in promoting axonal regeneration through glial scars after spinal cord injury.
Collapse
Affiliation(s)
- WESTLEY B. NOLIN
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
- Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York
| | - JAIME EMMETSBERGER
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
- Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York
| | - NOREEN BUKHARI
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
- Program in Neurosciences, Stony Brook University, Stony Brook, New York
| | - YAN ZHANG
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York
| | - JOEL M. LEVINE
- Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York
- Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York
- Program in Neurosciences, Stony Brook University, Stony Brook, New York
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York
| | - STELLA E. TSIRKA
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
- Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York
- Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York
- Program in Neurosciences, Stony Brook University, Stony Brook, New York
| |
Collapse
|
209
|
White RE, Jakeman LB. Don't fence me in: harnessing the beneficial roles of astrocytes for spinal cord repair. Restor Neurol Neurosci 2008; 26:197-214. [PMID: 18820411 PMCID: PMC2825119] [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: 05/26/2023]
Abstract
Astrocytes comprise a heterogeneous cell population that plays a complex role in repair after spinal cord injury. Reactive astrocytes are major contributors to the glial scar that is a physical and chemical barrier to axonal regeneration. Yet, consistent with a supportive role in development, astrocytes secrete neurotrophic factors and protect neurons and glia spared by the injury. In development and after injury, local cues are modulators of astrocyte phenotype and function. When multipotent cells are transplanted into the injured spinal cord, they differentiate into astrocytes and other glial cells as opposed to neurons, which is commonly viewed as a challenge to be overcome in developing stem cell technology. However, several examples show that astrocytes provide support and guidance for axonal growth and aid in improving functional recovery after spinal cord injury. Notably, transplantation of astrocytes of a developmentally immature phenotype promotes tissue sparing and axonal regeneration. Furthermore, interventions that enhance endogenous astrocyte migration or reinvasion of the injury site result in greater axonal growth. These studies demonstrate that astrocytes are dynamic, diverse cells that have the capacity to promote axon growth after injury. The ability of astrocytes to be supportive of recovery should be exploited in devising regenerative strategies.
Collapse
Affiliation(s)
- Robin E. White
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH, USA
- The Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Lyn B. Jakeman
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH, USA
- The Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
210
|
Fumagalli F, Madaschi L, Brenna P, Caffino L, Marfia G, Di Giulio AM, Racagni G, Gorio A. Single exposure to erythropoietin modulates Nerve Growth Factor expression in the spinal cord following traumatic injury: Comparison with methylprednisolone. Eur J Pharmacol 2008; 578:19-27. [DOI: 10.1016/j.ejphar.2007.09.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 08/31/2007] [Accepted: 09/24/2007] [Indexed: 11/30/2022]
|
211
|
Giger RJ, Venkatesh K, Chivatakarn O, Raiker SJ, Robak L, Hofer T, Lee H, Rader C. Mechanisms of CNS myelin inhibition: evidence for distinct and neuronal cell type specific receptor systems. Restor Neurol Neurosci 2008; 26:97-115. [PMID: 18820405 PMCID: PMC7259427] [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: 05/26/2023]
Abstract
Following injury to the adult mammalian central nervous system, regenerative growth of severed axons is very limited. The lack of neuronal repair is often associated with significant functional deficits, and depending on the severity of injury, may result in permanent paralysis distal to the site of injury. A detailed understanding of the molecular mechanisms that limit neuronal growth in the injured spinal cord is an important step toward the development of specific strategies aimed at restoring functional connectivity lost as a consequence of injury. While rapid progress is being made in defining the molecular identity of CNS growth inhibitory constituents, comparatively little is known about their receptors and downstream signaling mechanisms. Emerging new evidence suggests that the mechanisms for myelin inhibition are likely to be complex, involving multiple and distinct receptor systems that may operate in a redundant manner. Furthermore, the relative contribution of a specific ligand-receptor system to bring about growth inhibition may greatly vary among different neuronal cell types. Myelin-associated glycoprotein (MAG), for example, employs different mechanisms to inhibit neurite outgrowth of cerebellar, sensory, and retinal ganglion neurons in vitro. Nogo-A harbors distinct growth inhibitory regions, which employ different signaling mechanisms. The Nogo-66 receptor 1 (NgR1), a shared ligand binding component in a receptor complex for Nogo-66, MAG, and OMgp, participates in neuronal growth cone collapse to acutely presented myelin inhibitors, but is dispensable for longitudinal neurite outgrowth inhibition on substrate-bound Nogo-66, MAG, OMgp, or crude CNS myelin in vitro. Consistent with the idea of cell-type specific mechanisms for myelin inhibition, different types of CNS neurons possess very different regenerative capacities and respond differently to experimental treatment strategies in vivo. We speculate that differences in regenerative axonal growth among different fiber systems are a reflection of their intrinsic ability to elongate axons and their distinct cell surface receptor profiles to respond to the growth inhibitory extracellular milieu. The existence of cell type specific mechanisms to impair regenerative axonal growth in the CNS may have important implications for the development of treatment strategies. Depending on the fiber tract injured, different ligand-receptor systems may need to be targeted in order to elicit robust and long-distance regenerative axonal growth.
Collapse
Affiliation(s)
- Roman J Giger
- Center for Neural Development and Disorder, Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | | | | | | | | | | | | | |
Collapse
|
212
|
Jin Y, Ziemba KS, Smith GM. Axon growth across a lesion site along a preformed guidance pathway in the brain. Exp Neurol 2007; 210:521-30. [PMID: 18261727 DOI: 10.1016/j.expneurol.2007.11.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 11/30/2007] [Accepted: 11/30/2007] [Indexed: 11/16/2022]
Abstract
Our previous studies showed that axonal outgrowth from dorsal root ganglia (DRG) transplants in the adult rat brain could be directed toward a specific target location using a preformed growth-supportive pathway. This pathway induced axon growth within the corpus callosum across the midline to the opposite hemisphere. In this study, we examined whether such pathways would also support axon growth either through or around a lesion of the corpus callosum. Pathways expressing GFP, NGF, or FGF2/NGF were set up by multiple injections of adenovirus along the corpus callosum. Each pathway included the transplantation site in the left corpus callosum, 2.8 mm away from the midline, and a target site in the right corpus callosum, 2.5 mm from the midline. At the same time, a 1 mm lesion was made through the corpus callosum at the midline in an anteroposterior direction. A group of control animals received lesions and Ad-NGF injections only at the transplant and target sites, without a bridging pathway. DRG cell suspensions from postnatal day 1 or 2 rats were injected at the transplantation site three to four days later. Two weeks after transplantation, brain sections were stained using an anti-CGRP antibody. The CGRP+ axons were counted at 0.5 mm and 1.5 mm from the lesion site in both hemispheres. Few axons grew past the lesion in animals with control pathways, but there was robust axon growth across the lesion site in the FGF2/NGF and NGF-expressing pathways. This study indicated that preformed NGF and combination guidance pathways support more axon growth past a lesion in the adult mammalian brain.
Collapse
Affiliation(s)
- Ying Jin
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA.
| | | | | |
Collapse
|
213
|
Sandrow HR, Shumsky JS, Amin A, Houle JD. Aspiration of a cervical spinal contusion injury in preparation for delayed peripheral nerve grafting does not impair forelimb behavior or axon regeneration. Exp Neurol 2007; 210:489-500. [PMID: 18295206 DOI: 10.1016/j.expneurol.2007.11.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 11/21/2007] [Accepted: 11/26/2007] [Indexed: 01/17/2023]
Abstract
A peripheral nerve graft model was used to examine axonal growth after a unilateral cervical (C) contusion injury in adult rats and to determine if manipulation of an injury site prior to transplantation affects spontaneous behavioral recovery. After a short delay (7 d) the epicenter of a C4 contusion was exposed and aspirated without harming the cavity walls followed by apposition with one end of a pre-degenerated tibial nerve to the rostral cavity wall. After a longer delay (28 d) the aspirated cavity was treated with GDNF to promote regeneration by chronically injured neurons. In both groups forelimb and hindlimb locomotor scores decreased significantly 2 d after lesion site manipulation, but by 7 d, the forelimb score was not different from the pre-manipulation score. There was no significant difference in grid walking or grip strength scores for the affected forelimb in either group 7 d after contusion vs. 7 d after manipulation. Over 1500 brain stem and propriospinal neurons grew axons into the graft with either delay. These results demonstrate that a contusion injury site can be manipulated prior to transplantation without causing long-lasting forelimb or hindlimb behavioral deficits and that peripheral nerve grafts support axonal growth after acute or chronic contusion injury.
Collapse
Affiliation(s)
- Harra R Sandrow
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | | | | | | |
Collapse
|
214
|
Combined treatment using peripheral nerve graft and FGF-1: changes to the glial environment and differential macrophage reaction in a complete transected spinal cord. Neurosci Lett 2007; 433:163-9. [PMID: 18291581 DOI: 10.1016/j.neulet.2007.11.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Revised: 11/07/2007] [Accepted: 11/21/2007] [Indexed: 11/21/2022]
Abstract
We used a complete spinal cord transection model in which the T8 spinal segment was removed to study the effect of combined treatment of peripheral nerve graft and application of FGF-1 on the glial environment. The combined treatment resulted in reduced astrocytic glial scarring, reactive macrophage gliosis, and inhibitory proteoglycan in the back-degenerated white matter tract. While the macrophage activities in the back-degenerative tract were down-regulated, those in the grafted peripheral nerves and in the distal Wallerian degenerative tracts were not. We concluded that the combined treatment changed the glial environment in the back-degenerative tract, and differentially regulated the macrophage activities in the system, in favor of CNS regeneration.
Collapse
|
215
|
Hamel MG, Ajmo JM, Leonardo CC, Zuo F, Sandy JD, Gottschall PE. Multimodal signaling by the ADAMTSs (a disintegrin and metalloproteinase with thrombospondin motifs) promotes neurite extension. Exp Neurol 2007; 210:428-40. [PMID: 18178186 DOI: 10.1016/j.expneurol.2007.11.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 11/01/2007] [Accepted: 11/16/2007] [Indexed: 01/03/2023]
Abstract
Aggregating proteoglycans (PG) bearing chondroitin sulfate (CS) side chains associate with hyaluronan and various secreted proteins to form a complex of extracellular matrix (ECM) that inhibits neural plasticity in the central nervous system (CNS). Chondroitinase treatment depletes PGs of their CS side chains and enhances neurite extension. Increasing evidence from in vivo models indicates that proteolytic cleavage of the PG core protein by members of the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of glutamyl-endopeptidases also promotes neural plasticity. The purpose of this study was to determine whether proteolytic action of the ADAMTSs influences neurite outgrowth in cultured neurons. Transfection of primary rat neurons with ADAMTS4 cDNA induced longer neurites, whether the neurons were grown on a monolayer of astrocytes that secrete inhibitory PGs or on laminin/poly-L-lysine substrate alone. Similar results were found when neurons were transfected with a construct encoding a proteolytically inactive, point mutant of ADAMTS4. Addition of recombinant ADAMTS4 or ADAMTS5 protein to immature neuronal cultures also enhanced neurite extension in a dose-dependent manner, an effect demonstrated to be dependent on the activation of MAP ERK1/2 kinase. These results suggest that ADAMTS4 enhances neurite outgrowth via a mechanism that does not require proteolysis but is dependent on activation of the MAP kinase cascade. Thus a model to illustrate multimodal ADAMTS activity would entail proteolysis of CS-bearing PGs to create a loosened matrix environment more favorable for neurite outgrowth, and enhanced neurite outgrowth directly stimulated by ADAMTS signaling at the cell surface.
Collapse
Affiliation(s)
- Michelle G Hamel
- University of South Florida College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612-4799, USA
| | | | | | | | | | | |
Collapse
|
216
|
García-Alías G, Lin R, Akrimi SF, Story D, Bradbury EJ, Fawcett JW. Therapeutic time window for the application of chondroitinase ABC after spinal cord injury. Exp Neurol 2007; 210:331-8. [PMID: 18158149 DOI: 10.1016/j.expneurol.2007.11.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 11/08/2007] [Accepted: 11/08/2007] [Indexed: 11/17/2022]
Abstract
Rats with a crush in the dorsal funiculi of the C4 segment of the spinal cord were treated with chondroitinase ABC delivered to the lateral ventricle, receiving 6 intraventricular injections on alternate days. In order to investigate the time window of efficacy of chondroitinase, treatment was begun at the time of injury or after a 2, 4 or 7 days delay. Behavioural testing over 6 weeks showed that acutely treated animals showed improved skilled forelimb reaching compared to penicillinase controls. Forelimb contact placing recovered in treated animals but not controls, and gait analysis showed recovery towards normal forelimb stride length in treated animals but not controls. Chondroitinase-treated animals showed greater axon regeneration than controls. The treatment effect on contact placing, stride length and axon regeneration was not dependent on the timing of the start of treatment, but in skilled paw reaching acutely treated animals recovered better function. The area of chondroitinase ABC digestion visualized by stub antibody staining included widespread digestion around the lateral ventricles and partial digestion of cervical spinal cord white matter, but not grey matter.
Collapse
Affiliation(s)
- Guillermo García-Alías
- Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge CB2 0PY, UK
| | | | | | | | | | | |
Collapse
|
217
|
Vitellaro-Zuccarello L, Mazzetti S, Madaschi L, Bosisio P, Fontana E, Gorio A, De Biasi S. Chronic erythropoietin-mediated effects on the expression of astrocyte markers in a rat model of contusive spinal cord injury. Neuroscience 2007; 151:452-66. [PMID: 18065151 DOI: 10.1016/j.neuroscience.2007.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 11/02/2007] [Accepted: 11/06/2007] [Indexed: 12/11/2022]
Abstract
Using a standardized rat model of contusive spinal cord injury (SCI; [Gorio A, Gokmen N, Erbayraktar S, Yilmaz O, Madaschi L, Cichetti C, Di Giulio AM, Vardar E, Cerami A, Brines M (2002) Recombinant human erythropoietin counteracts secondary injury and markedly enhances neurological recovery from experimental spinal cord trauma. Proc Natl Acad Sci U S A 99:9450-9455]), we previously showed that the administration of recombinant human erythropoietin (rhEPO) improves both tissue sparing and locomotory outcome. In the present study, to better understand rhEPO-mediated effects on chronic astrocyte response to SCI in rat, we have used immunocytochemical methods combined with confocal and electron microscopy to investigate, 1 month after injury, the effects of a single rhEPO administration on the expression of a) aquaporin 4 (AQP4), the main astrocytic water channel implicated in edema development and resolution, and two molecules (dystrophin and syntrophin) involved in its membrane anchoring; b) glial fibrillary acidic protein (GFAP) and vimentin as markers of astrogliosis; c) chondroitin sulfate proteoglycans of the extracellular matrix which are upregulated after SCI and can inhibit axonal regeneration and influence neuronal and glial properties. Our results show that rhEPO administration after SCI modifies astrocytic response to injury by increasing AQP4 immunoreactivity in the spinal cord, but not in the brain, without apparent modifications of dystrophin and syntrophin distribution. Attenuation of astrogliosis, demonstrated by the semiquantitative analysis of GFAP labeling, was associated with a reduction of phosphacan/RPTP zeta/beta, whereas the levels of lecticans remained unchanged. Finally, the relative volume of a microvessel fraction was significantly increased, indicating a pro-angiogenetic or a vasodilatory effect of rhEPO. These changes were consistently associated with remarkable reduction of lesion size and with improvement in tissue preservation and locomotor recovery, confirming previous observations and underscoring the potentiality of rhEPO for the therapeutic management of SCI.
Collapse
Affiliation(s)
- L Vitellaro-Zuccarello
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Università degli Studi di Milano, Via Celoria, 26 I-20133 Milano, Italy.
| | | | | | | | | | | | | |
Collapse
|
218
|
Benowitz LI, Yin Y. Combinatorial treatments for promoting axon regeneration in the CNS: strategies for overcoming inhibitory signals and activating neurons' intrinsic growth state. Dev Neurobiol 2007; 67:1148-65. [PMID: 17514713 DOI: 10.1002/dneu.20515] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In general, neurons in the mature mammalian central nervous system (CNS) are unable to regenerate injured axons, and neurons that remain uninjured are unable to form novel connections that might compensate for ones that have been lost. As a result of this, victims of CNS injury, stroke, or certain neurodegenerative diseases are unable to fully recover sensory, motor, cognitive, or autonomic functions. Regenerative failure is related to a host of inhibitory signals associated with the extracellular environment and with the generally low intrinsic potential of mature CNS neurons to regenerate. Most research to date has focused on extrinsic factors, particularly the identification of inhibitory proteins associated with myelin, the perineuronal net, glial cells, and the scar that forms at an injury site. However, attempts to overcome these inhibitors have resulted in relatively limited amounts of CNS regeneration. Using the optic nerve as a model system, we show that with appropriate stimulation, mature neurons can revert to an active growth state and that when this occurs, the effects of overcoming inhibitory signals are enhanced dramatically. Similar conclusions are emerging from studies in other systems, pointing to a need to consider combinatorial treatments in the clinical setting.
Collapse
Affiliation(s)
- Larry I Benowitz
- Laboratories for Neuroscience Research in Neurosurgery and Neurobiology Program, Children's Hospital, Boston, Massachusetts, USA.
| | | |
Collapse
|
219
|
Tester NJ, Howland DR. Chondroitinase ABC improves basic and skilled locomotion in spinal cord injured cats. Exp Neurol 2007; 209:483-96. [PMID: 17936753 DOI: 10.1016/j.expneurol.2007.07.019] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 07/21/2007] [Accepted: 07/25/2007] [Indexed: 01/08/2023]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are upregulated in the central nervous system following injury. Chondroitin sulfate glycosaminoglycan (CS GAG) side chains substituted on this family of molecules contribute to the limited functional recovery following injury by restricting axonal growth and synaptic plasticity. In the current study, the effects of degrading CS GAGs with Chondroitinase ABC (Ch'ase ABC) in the injured spinal cords of adult cats were assessed. Three groups were evaluated for 5 months following T10 hemisections: lesion-only, lesion+control, and lesion+Ch'ase ABC. Intraspinal control and Ch'ase ABC treatments to the lesion site began immediately after injury and continued every other day, for a total of 15 treatments, using an injectable port system. Delivery and in vivo cleavage were verified anatomically in a subset of cats across the treatment period. Recovery of skilled locomotion (ladder, peg, and beam) was significantly accelerated, on average, by >3 weeks in Ch'ase ABC-treated cats compared to controls. Ch'ase ABC-treated cats also showed greater recovery of specific skilled locomotor features including intralimb movement patterns and significantly greater paw placement onto pegs. Although recovery of basic locomotion (bipedal treadmill and overground) was not accelerated, intralimb movement patterns were more normal in the Ch'ase ABC-treated cats. Qualitative assessment of serotonergic immunoreactivity also suggested that Ch'ase ABC treatment enhanced plasticity. Finally, analyses using fluorophore-assisted carbohydrate electrophoresis (FACE) indicate CS GAG content is similar in cat and human. These findings show, for the first time, that intraspinal cleavage of CS GAGs can enhance recovery of function following spinal cord injury in large animals with sophisticated motor behaviors and axonal growth requirements similar to those encountered in humans.
Collapse
Affiliation(s)
- Nicole J Tester
- Malcom Randall Veterans Affairs Medical Center, Gainesville, FL 32608, USA
| | | |
Collapse
|
220
|
Gris P, Tighe A, Levin D, Sharma R, Brown A. Transcriptional regulation of scar gene expression in primary astrocytes. Glia 2007; 55:1145-55. [PMID: 17597120 DOI: 10.1002/glia.20537] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The failure of the adult injured spinal cord to support axonal regeneration is in part attributed to the glial scar. Reactive astrocytes constitute a major cellular component of the glial scar and are heterogeneous with respect to the extracellular matrix proteins that they secrete. Astrocytes may produce antiregenerative molecules such as chondroitin sulphate proteoglycans (CSPGs) or proregenerative molecules such as laminin and fibronectin. While many different CSPGs are expressed after spinal cord injury (SCI) they all rely on the same enzymes, xylosyltransferase-I and -II (XT-I, XT-II) and chondroitin 4-sulfotransferase (C4ST) to add the repulsive chondroitin sulfate side chains to their core proteins. We show that XT-I, XT-II, and C4ST are part of a CSPG biosynthetic gene (CBG) battery. Using primary astrocyte cultures and quantitative PCR we demonstrate that TGFbeta2, PDGF, and IL-6 induce the expression of CBGs, laminin and fibronectin by several-fold. We further show that over-expression of the transcription factor SOX9 also strongly induces the expression of CBGs but does not increase the expression of laminin or fibronectin. Correspondingly, SOX9 knock-down in primary astrocytes causes a decrease in CBG and an increase in laminin and fibronectin mRNA levels. Finally, we show that the in vivo expression profiles of TGFbeta2, PDGF, IL-6, and SOX9 are consistent with their potential roles in differentially regulating CBGs, laminin and fibronectin gene expression in the injured spinal cord. This work suggests that SOX9 levels may be pivotal in determining the balance of pro- and anti-regenerative extracellular matrix molecules produced by astrocytes.
Collapse
Affiliation(s)
- Paul Gris
- The Spinal Cord Injury Team, BioTherapeutics Research Group, Robarts Research Institute, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
221
|
Sotogaku N, Tully SE, Gama CI, Higashi H, Tanaka M, Hsieh-Wilson LC, Nishi A. Activation of phospholipase C pathways by a synthetic chondroitin sulfate-E tetrasaccharide promotes neurite outgrowth of dopaminergic neurons. J Neurochem 2007; 103:749-60. [PMID: 17680989 DOI: 10.1111/j.1471-4159.2007.04849.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In dopaminergic neurons, chondroitin sulfate (CS) proteoglycans play important roles in neuronal development and regeneration. However, due to the complexity and heterogeneity of CS, the precise structure of CS with biological activity and the molecular mechanisms underlying its influence on dopaminergic neurons are poorly understood. In this study, we investigated the ability of synthetic CS oligosaccharides and natural polysaccharides to promote the neurite outgrowth of mesencephalic dopaminergic neurons and the signaling pathways activated by CS. CS-E polysaccharide, but not CS-A, -C or -D polysaccharide, facilitated the neurite outgrowth of dopaminergic neurons at CS concentrations within the physiological range. The stimulatory effect of CS-E polysaccharide on neurite outgrowth was completely abolished by its digestion into disaccharide units with chondroitinase ABC. Similarly to CS-E polysaccharide, a synthetic tetrasaccharide displaying only the CS-E sulfation motif stimulated the neurite outgrowth of dopaminergic neurons, whereas a CS-E disaccharide or unsulfated tetrasaccharide had no effect. Analysis of the molecular mechanisms revealed that the action of the CS-E tetrasaccharide was mediated through midkine-pleiotrophin/protein tyrosine phosphatase zeta and brain-derived neurotrophic factor/tyrosine kinase B receptor pathways, followed by activation of the two intracellular phospholipase C (PLC) signaling cascades: PLC/protein kinase C and PLC/inositol 1,4,5-triphosphate/inositol 1,4,5-triphosphate receptor signaling leading to intracellular Ca(2+) concentration-dependent activation of Ca(2+)/calmodulin-dependent kinase II and calcineurin. These results indicate that a specific sulfation motif, in particular the CS-E tetrasaccharide unit, represents a key structural determinant for activation of midkine, pleiotrophin and brain-derived neurotrophic factor-mediated signaling, and is required for the neuritogenic activity of CS in dopaminergic neurons.
Collapse
Affiliation(s)
- Naoki Sotogaku
- Department of Pharmacology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
222
|
Shelke SV, Gao GP, Mesch S, Gäthje H, Kelm S, Schwardt O, Ernst B. Synthesis of sialic acid derivatives as ligands for the myelin-associated glycoprotein (MAG). Bioorg Med Chem 2007; 15:4951-65. [PMID: 17507233 DOI: 10.1016/j.bmc.2007.04.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Revised: 04/11/2007] [Accepted: 04/20/2007] [Indexed: 11/27/2022]
Abstract
The trisaccharide substructure 13 of the ganglioside GQ1balpha shows a remarkable affinity for the myelin-associated glycoprotein (MAG). In the search for structurally simplified and pharmacokinetically improved mimics of 13, sialosides with modifications at the reducing and non-reducing end were synthesized. The biological evaluation of mimics 12a-o was performed in a competitive target-based assay. It was found that the relative inhibitory potency (rIP) of antagonist 12h was enhanced by more than 1000-fold in comparison to the reference trisaccharide 13, despite the former having a much simpler structure. In addition, the sialic acid derivatives, for example, 12h, have clearly improved pharmacokinetic properties due to the presence of aromatic moieties, a lower molecular weight, and a reduced number of polar hydroxy functions compared to the reference compound 13.
Collapse
Affiliation(s)
- Sachin V Shelke
- Institute of Molecular Pharmacy, Pharmacenter, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
223
|
Fitch MT, Silver J. CNS injury, glial scars, and inflammation: Inhibitory extracellular matrices and regeneration failure. Exp Neurol 2007; 209:294-301. [PMID: 17617407 PMCID: PMC2268907 DOI: 10.1016/j.expneurol.2007.05.014] [Citation(s) in RCA: 745] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Accepted: 05/22/2007] [Indexed: 11/20/2022]
Abstract
Spinal cord and brain injuries lead to complex cellular and molecular interactions within the central nervous system in an attempt to repair the initial tissue damage. Many studies have illustrated the importance of the glial cell response to injury, and the influences of inflammation and wound healing processes on the overall morbidity and permanent disability that result. The abortive attempts of neuronal regeneration after spinal cord injury are influenced by inflammatory cell activation, reactive astrogliosis and the production of both growth promoting and inhibitory extracellular molecules. Despite the historical perspective that the glial scar was a mechanical barrier to regeneration, inhibitory molecules in the forming scar and methods to overcome them have suggested molecular modification strategies to allow neuronal growth and functional regeneration. Unlike myelin associated inhibitory molecules, which remain at largely static levels before and after central nervous system trauma, inhibitory extracellular matrix molecules are dramatically upregulated during the inflammatory stages after injury providing a window of opportunity for the delivery of candidate therapeutic interventions. While high dose methylprednisolone steroid therapy alone has not proved to be the solution to this difficult clinical problem, other strategies for modulating inflammation and changing the make up of inhibitory molecules in the extracellular matrix are providing robust evidence that rehabilitation after spinal cord and brain injury has the potential to significantly change the outcome for what was once thought to be permanent disability.
Collapse
Affiliation(s)
- Michael T Fitch
- Department of Emergency Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
224
|
Shen LH, Li Y, Chen J, Cui Y, Zhang C, Kapke A, Lu M, Savant-Bhonsale S, Chopp M. One-year follow-up after bone marrow stromal cell treatment in middle-aged female rats with stroke. Stroke 2007; 38:2150-6. [PMID: 17525391 DOI: 10.1161/strokeaha.106.481218] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND PURPOSE We sought to evaluate the long-term effects of bone marrow stromal cell (BMSC) treatment on retired breeder rats with stroke. METHODS Female retired breeder rats were subjected to 2-hour middle cerebral artery occlusion (MCAO) followed by an injection of 2 x 10(6) male BMSCs (n=8) or phosphate-buffered saline (n=11) into the ipsilateral internal carotid artery at 1 day after stroke. The rats were humanely killed 1 year later. Functional tests, in situ hybridization, and histochemical and immunohistochemical staining were performed. RESULTS Significant recovery of neurological deficits was found in BMSC-treated rats beginning 2 weeks after cell injection compared with control animals. The beneficial effects of cell transplantation persisted for at least 1 year (P<0.01). In situ hybridization for the Y chromosome showed that donor cells survived in the brains of recipient rats, among which 22.3+/-1.95% of cells expressed the astrocyte marker glial fibrillary acidic protein, 16.8+/-2.13% expressed the neuronal marker microtubule-associated protein 2, and 5.5+/-0.42% and <1% of cells colocalized with the microglial marker IB4 and the endothelial cell marker von Willebrand factor, respectively. Only very few BMSCs, however, were found in peripheral organs such as the heart, lung, liver, spleen, and kidney in recipient rats. BMSCs significantly reduced axonal loss (P<0.01), the thickness of the lesion scar wall (P<0.01), and the number of Nogo-A-positive cells (P<0.05) along the scar border; meanwhile, synaptophysin expression (P<0.05) was significantly increased in BMSC-treated ischemic brains compared with control untreated brains. CONCLUSIONS The beneficial effects of BMSCs on ischemic brain tissue persisted for at least 1 year. Most surviving BMSCs were present in the ischemic brain, but very few were found in other organs. The long-term improvement in functional outcome may be related to the structural and molecular changes induced by BMSCs.
Collapse
Affiliation(s)
- Li Hong Shen
- Department of Neurology, Henry Ford Hospital, E&R 3056, Detroit, MI 48202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Li HP, Homma A, Sango K, Kawamura K, Raisman G, Kawano H. Regeneration of nigrostriatal dopaminergic axons by degradation of chondroitin sulfate is accompanied by elimination of the fibrotic scar and glia limitans in the lesion site. J Neurosci Res 2007; 85:536-47. [PMID: 17154415 DOI: 10.1002/jnr.21141] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Chondroitin sulfate increases around a lesion site after central nervous system injury and is believed to be an impediment to axonal regeneration, because administration of chondroitinase ABC, a chondroitin sulfate-degrading enzyme, promotes axonal regeneration of central neurons. To examine the physiological role of chondroitin sulfate up-regulation after injury, the nigrostriatal dopaminergic axons were unilaterally transected in mice, and chondroitinase ABC was then injected into the lesion site. In mice transected only, tyrosine hydroxylase-immunoreactive axons did not extend across the lesion at 1 or 2 weeks after the transection. Immunoreactivities of chondroitin sulfate side chains and core protein of NG2 proteoglycan increased in and around the lesion site, and a fibrotic scar containing type IV collagen deposits developed in the lesion center. In contrast, in mice transected and treated with chondroitinase ABC, numerous tyrosine hydroxylase-immunoreactive axons were regenerated across the lesion at 1 and 2 weeks after the transection. In these animals, chondroitin sulfate immunoreactivity remarkably decreased, and immunoreactivity of 2B6 antibody, which recognizes the stub of degraded chondroitin sulfate side chains, was enhanced. Furthermore, the formation of a fibrotic scar and a glia limitans that surrounds the former was completely prevented, although type IV collagen immunoreactivity remained in newly formed blood capillaries around the lesion site. We discuss the question of whether the chondroitin sulfate is acting as a direct inhibitor of axonal regeneration or whether the observed changes are due to a prevention of the fibrotic scar formation and a rearrangement of astrocytic membranes.
Collapse
Affiliation(s)
- Hong-Peng Li
- Department of Developmental Morphology, Tokyo Metropolitan Institute for Neuroscience, Fuchu, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
226
|
Crespo D, Asher RA, Lin R, Rhodes KE, Fawcett JW. How does chondroitinase promote functional recovery in the damaged CNS? Exp Neurol 2007; 206:159-71. [PMID: 17572406 DOI: 10.1016/j.expneurol.2007.05.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 04/28/2007] [Accepted: 05/02/2007] [Indexed: 12/22/2022]
Abstract
A number of recent studies have established that the bacterial enzyme chondroitinase ABC promotes functional recovery in the injured CNS. The issue of how it works is rarely addressed, however. The effects of the enzyme are presumed to be due to the degradation of inhibitory chondroitin sulphate GAG chains. Here we review what is known about the composition, structure and distribution of the extracellular matrix in the CNS, and how it changes in response to injury. We summarize the data pertaining to the ability of chondroitinase to promote functional recovery, both in the context of axon regeneration and the reactivation of plasticity. We also present preliminary data on the persistence of the effects of the enzyme in vivo, and its hyaluronan-degrading activity in CNS homogenates in vitro. We then consider precisely how the enzyme might influence functional recovery in the CNS. The ability of chondroitinase to degrade hyaluronan is likely to result in greater matrix disruption than the degradation of chondroitin sulphate alone.
Collapse
Affiliation(s)
- Dámaso Crespo
- Cambridge University Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge, CB2 2PY, UK
| | | | | | | | | |
Collapse
|
227
|
Massey JM, Amps J, Viapiano MS, Matthews RT, Wagoner MR, Whitaker CM, Alilain W, Yonkof AL, Khalyfa A, Cooper NGF, Silver J, Onifer SM. Increased chondroitin sulfate proteoglycan expression in denervated brainstem targets following spinal cord injury creates a barrier to axonal regeneration overcome by chondroitinase ABC and neurotrophin-3. Exp Neurol 2007; 209:426-45. [PMID: 17540369 PMCID: PMC2270474 DOI: 10.1016/j.expneurol.2007.03.029] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2007] [Revised: 03/24/2007] [Accepted: 03/27/2007] [Indexed: 01/11/2023]
Abstract
Increased chondroitin sulfate proteoglycan (CSPG) expression in the vicinity of a spinal cord injury (SCI) is a primary participant in axonal regeneration failure. However, the presence of similar increases of CSPG expression in denervated synaptic targets well away from the primary lesion and the subsequent impact on regenerating axons attempting to approach deafferented neurons have not been studied. Constitutively expressed CSPGs within the extracellular matrix and perineuronal nets of the adult rat dorsal column nuclei (DCN) were characterized using real-time PCR, Western blot analysis and immunohistochemistry. We show for the first time that by 2 days and through 3 weeks following SCI, the levels of NG2, neurocan and brevican associated with reactive glia throughout the DCN were dramatically increased throughout the DCN despite being well beyond areas of trauma-induced blood brain barrier breakdown. Importantly, regenerating axons from adult sensory neurons microtransplanted 2 weeks following SCI between the injury site and the DCN were able to regenerate rapidly within white matter (as shown previously by Davies et al. [Davies, S.J., Goucher, D.R., Doller, C., Silver, J., 1999. Robust regeneration of adult sensory axons in degenerating white matter of the adult rat spinal cord. J. Neurosci. 19, 5810-5822]) but were unable to enter the denervated DCN. Application of chondroitinase ABC or neurotrophin-3-expressing lentivirus in the DCN partially overcame this inhibition. When the treatments were combined, entrance by regenerating axons into the DCN was significantly augmented. These results demonstrate both an additional challenge and potential treatment strategy for successful functional pathway reconstruction after SCI.
Collapse
Affiliation(s)
- James M. Massey
- M.D./Ph.D. Program, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Jeremy Amps
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Mariano S. Viapiano
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, 06520
| | - Russell. T. Matthews
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, 06520
| | - Michelle R. Wagoner
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Christopher M. Whitaker
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Warren Alilain
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Alicia L. Yonkof
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Abdelnaby Khalyfa
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Nigel G. F. Cooper
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
- Address for correspondence, proofs, and reprint requests: Stephen M. Onifer, Ph.D., Spinal Cord and Brain Injury Research Center, University of Kentucky, BBSRB B365, 741 South Limestone Street, Lexington, KY, 40536-0509, U.S.A., TELEPHONE: (859) 323-5226, FAX: (859) 257-5737, EMAIL:
| | - Stephen M. Onifer
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| |
Collapse
|
228
|
Nyatia E, Lang DM. Localisation and expression of a myelin associated neurite inhibitor, Nogo-A and its receptor Nogo-receptor by mammalian CNS cells. Res Vet Sci 2007; 83:287-301. [PMID: 17428512 DOI: 10.1016/j.rvsc.2007.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Revised: 11/17/2006] [Accepted: 01/21/2007] [Indexed: 10/23/2022]
Abstract
Axon regeneration failure in the adult mammalian central nervous system (CNS) is partly due to inhibitory molecules associated with myelin. The Nogo receptor (NgR) plays a role in this process through an extraordinary degree of cross reactivity with three structurally unrelated myelin-associated inhibitory ligands namely; Nogo-A, myelin associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp). The major aim of the study was to investigate and explore the cellular localisation and expression pattern of NgR and Nogo-A in the mammalian nervous system. We therefore generated a rabbit polyclonal anti-NgR antibody from the leucine rich repeat (LRR) No. 9 domain of the NgR polypeptide chain. Together with a commercially available polyclonal antibody specific for NgR, and in conjunction with double labeling immunofluorescence methods on cryosections and cell cultures, NgR immunoreactivity was observed in the CNS and dorsal root ganglia (DRG). In cellular populations, it was confined to neuronal cell bodies and their processes. NgR was also localised on the surface of extending DRG intact axons and growth cones in live staining experiments. Nogo-A, a member of the reticulon family protein, was widely distributed in the mammalian brain, spinal cord, and DRG. Intense Nogo-A immunoreactivity was also detected in oligodendrocyte cell bodies and their myelin sheaths in nerve fibre tracts of the CNS. Furthermore, numerous populations of neurons in the brain and spinal cord expressed Nogo-A to a variable extent in their cell bodies and neurites, suggesting additional, as-yet-unknown, functions of this protein. These results confirm results obtained by other researchers with different sets of antibodies. However, they also raise the question of the mechanism and circumstances under which NgR interacts with Nogo-A, as the latter appears to be confined to the cytoplasm and can therefore not be expected to bind NgR on the axon surface.
Collapse
Affiliation(s)
- E Nyatia
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa.
| | | |
Collapse
|
229
|
Zurn AD, Bandtlow CE. Regeneration failure in the CNs: cellular and molecular mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:54-76. [PMID: 16955704 DOI: 10.1007/0-387-30128-3_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Anne D Zurn
- Department of Experimental Surgery, Lausanne University Hospital, Faculty of Biology and Medicine, Switzerland
| | | |
Collapse
|
230
|
Faissner A, Heck N, Dobbertin A, Garwood J. DSD-1-Proteoglycan/Phosphacan and Receptor Protein Tyrosine Phosphatase-Beta Isoforms during Development and Regeneration of Neural Tissues. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:25-53. [PMID: 16955703 DOI: 10.1007/0-387-30128-3_3] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Interactions between neurons and glial cells play important roles in regulating key events of development and regeneration of the CNS. Thus, migrating neurons are partly guided by radial glia to their target, and glial scaffolds direct the growth and directional choice of advancing axons, e.g., at the midline. In the adult, reactive astrocytes and myelin components play a pivotal role in the inhibition of regeneration. The past years have shown that astrocytic functions are mediated on the molecular level by extracellular matrix components, which include various glycoproteins and proteoglycans. One important, developmentally regulated chondroitin sulfate proteoglycan is DSD-1-PG/phosphacan, a glial derived proteoglycan which represents a splice variant of the receptor protein tyrosine phosphatase (RPTP)-beta (also known as PTP-zeta). Current evidence suggests that this proteoglycan influences axon growth in development and regeneration, displaying inhibitory or stimulatory effects dependent on the mode of presentation, and the neuronal lineage. These effects seem to be mediated by neuronal receptors of the Ig-CAM superfamily.
Collapse
Affiliation(s)
- Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University, Bochum, Germany
| | | | | | | |
Collapse
|
231
|
Deller T, Haas CA, Freiman TM, Phinney A, Jucker M, Frotscher M. Lesion-Induced Axonal Sprouting in the Central Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:101-21. [PMID: 16955706 DOI: 10.1007/0-387-30128-3_6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Injury or neuronal death often come about as a result of brain disorders. Inasmuch as the damaged nerve cells are interconnected via projections to other regions of the brain, such lesions lead to axonal loss in distal target areas. The central nervous system responds to deafferentation by means of plastic remodeling processes, in particular by inducing outgrowth of new axon collaterals from surviving neurons (collateral sprouting). These sprouting processes result in a partial reinnervation, new circuitry, and functional changes within the deafferented brain regions. Lesioning of the entorhinal cortex is an established model system for studying the phenomenon of axonal sprouting. Using this model system, it could be shown that the sprouting process respects the pre-existing lamination pattern of the deafferented fascia dentata, i. e., it is layer-specific. A variety of different molecules are involved in regulating this reorganization process (extracellular matrix molecules, cell adhesion molecules, transcription factors, neurotrophic factors, growth-associated proteins). It is proposed here that molecules of the extracellular matrix define the boundaries of the laminae following entorhinal lesioning and in so doing limit the sprouting process to the deafferented zone. To illustrate the role of axonal sprouting in disease processes, special attention is given to its significance for neurodegenerative disorders, particularly Alzheimer's disease (AD), and temporal lobe epilepsy. Finally, we discuss both the beneficial as well as disadvantageous functional implications of axonal sprouting for the injured organism in question.
Collapse
Affiliation(s)
- Thomas Deller
- Institute of Clinical Neuroanatomy, Johann Wolfgang Goether-University, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
232
|
Fawcett JW. The Glial Response to Injury and Its Role in the Inhibition of CNS Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:11-24. [PMID: 16955702 DOI: 10.1007/0-387-30128-3_2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
233
|
Busch SA, Silver J. The role of extracellular matrix in CNS regeneration. Curr Opin Neurobiol 2007; 17:120-7. [PMID: 17223033 DOI: 10.1016/j.conb.2006.09.004] [Citation(s) in RCA: 347] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Accepted: 09/25/2006] [Indexed: 10/23/2022]
Abstract
Chondroitin sulfate proteoglycans are the principal inhibitory component of glial scars, which form after damage to the adult central nervous system and act as a barrier to regenerating axons. Recent findings have furthered our understanding of the mechanisms that result in a failure of regeneration after spinal cord injury and suggest that a multipartite approach will be required to facilitate long-distance regeneration and functional recovery.
Collapse
Affiliation(s)
- Sarah A Busch
- Case Western Reserve University School of Medicine, Department of Neurosciences, 2109 Adelbert Road E-658, Cleveland, Ohio 44106, USA
| | | |
Collapse
|
234
|
Abstract
Neural recognition molecules were discovered and characterized initially for their functional roles in cell adhesion as regulators of affinity between cells and the extracellular matrix in vitro. They were then recognized as mediators or co-receptors which trigger signal transduction mechanisms affecting cell adhesion and de-adhesion. Their involvement in contact attraction and repulsion relies on cell-intrinsic properties that are modulated by the spatial contexts of their expression at particular stages of ontogenetic development, in synaptic plasticity and during regeneration after injury. The functional roles of recognition molecules in cell proliferation and migration, determination of developmental fate, growth cone guidance, and synapse formation, stabilization and modulation have been well documented not only by in vitro, but also by in vivo studies that have been greatly aided by generation of genetically altered mice. More recently, the functions of recognition molecules have been investigated under conditions of neural repair and manipulated using a broad range of genetic and pharmacological approaches to achieve a beneficial outcome. The principal aim of most therapeutically oriented approaches has been to neutralize inhibitory factors. However, less attention has been paid to enhancing repair by stimulating the stimulatory factors. When considering potential therapeutic strategies, it is worth considering that a single recognition molecule can possess domains that are conducive or repellent and that the spatial distribution of recognition molecules can determine the overall function: Recognition molecules may be repellent for neurite outgrowth when presented as barriers or steep-concentration gradients and conducive when presented as uniform substrates. The focus of this review will be on the more recent attempts to study the conducive mechanisms with the expectation that they may be able to tip the balance from a regeneration inhospitable to a hospitable environment. It is likely that a combination of the two principles, as multifactorial as each principle may be in itself, will be of therapeutic value in humans.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Universität Hamburg, Hamburg, Germany
| | | |
Collapse
|
235
|
Berglöf E, Plantman S, Johansson S, Strömberg I. Inhibition of proteoglycan synthesis affects neuronal outgrowth and astrocytic migration in organotypic cultures of fetal ventral mesencephalon. J Neurosci Res 2007; 86:84-92. [PMID: 17868150 DOI: 10.1002/jnr.21465] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Grafting fetal ventral mesencephalon has been utilized to alleviate the symptoms of Parkinson's disease. One obstacle in using this approach is the limited outgrowth from the transplanted dopamine neurons. Thus, it is important to evaluate factors that promote outgrowth from fetal dopamine neurons. Proteoglycans (PGs) are extracellular matrix molecules that modulate neuritic growth. This study was performed to evaluate the role of PGs in dopamine nerve fiber formation in organotypic slice cultures of fetal ventral mesencephalon. Cultures were treated with the PG synthesis inhibitor methyl-umbelliferyl-beta-D-xyloside (beta-xyloside) and analyzed using antibodies against tyrosine hydroxylase (TH) to visualize dopamine neurons, S100beta to visualize astrocytes, and neurocan to detect PGs. Two growth patterns of TH-positive outgrowth were observed: nerve fibers formed in the presence of astrocytes and nerve fibers formed in the absence of astrocytes. Treatment with beta-xyloside significantly reduced the distance of glial-associated TH-positive nerve fiber outgrowth but did not affect the length of the non-glial-associated nerve fibers. The addition of beta-xyloside shifted the nerve fiber growth pattern from being mostly glial-guided to being non-glial-associated, whereas the total amount of TH protein was not affected. Further, astrocytic migration and proliferation were impaired after beta-xyloside treatment, and levels of non-intact PG increased. beta-Xyloside treatment changed the distribution of neurocan in astrocytes, from being localized in vesicles to being diffusely immunoreactive in the processes. To conclude, inhibition of PG synthesis affects glial-associated TH-positive nerve fiber formation in ventral mesencephalic cultures, which might be an indirect effect of impaired astrocytic migration.
Collapse
Affiliation(s)
- Elisabet Berglöf
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | | | | |
Collapse
|
236
|
Pettigrew DB, Li YQ, Kuntz C, Crutcher KA. Global expression of NGF promotes sympathetic axonal growth in CNS white matter but does not alter its parallel orientation. Exp Neurol 2007; 203:95-109. [PMID: 16989811 PMCID: PMC2638215 DOI: 10.1016/j.expneurol.2006.07.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Revised: 07/18/2006] [Accepted: 07/26/2006] [Indexed: 11/30/2022]
Abstract
Axonal regeneration is normally limited after injuries to CNS white matter. Infusion of neurotrophins has been successful in promoting regenerative growth through injured white matter but this growth generally fails to extend beyond the infusion site. These observations are consistent with a chemotropic effect of these factors on axonal growth and support the prevailing view that neurotrophin-induced axonal regeneration requires the use of gradients, i.e., gradually increasing neurotrophin levels along the target fiber tract. To examine the potential of global overexpression of neurotrophins to promote, and/or modify the orientation of, regenerative axonal growth within white matter, we grafted nerve growth factor (NGF) responsive neurons into the corpus callosum of transgenic mice overexpressing NGF throughout the CNS under control of the promoter for glial fibrillary acidic protein. One week later, glial fibrillary acidic protein and chondroitin sulfate proteoglycan immunoreactivity increased within injured white matter around the grafts. NGF levels were significantly higher in the brains of transgenic compared with non-transgenic mice and further elevated within injury sites compared with the homotypic region of the non-injured side. Although there was minimal outgrowth from neurons grafted into non-transgenic mice, extensive parallel axonal regeneration had occurred within the corpus callosum up to 1.5 mm beyond the astrogliotic scar (the site of maximum NGF expression) in transgenic mice. These results demonstrate that global overexpression of neurotrophins does not override the constraints limiting regenerative growth to parallel orientations and suggest that such factors need not be presented as positive gradients to promote axonal regeneration within white matter.
Collapse
MESH Headings
- Animals
- Astrocytes/cytology
- Astrocytes/physiology
- Axotomy
- Brain Injuries/metabolism
- Brain Injuries/physiopathology
- Brain Injuries/therapy
- Brain Injury, Chronic/metabolism
- Brain Injury, Chronic/physiopathology
- Brain Injury, Chronic/therapy
- Central Nervous System/cytology
- Central Nervous System/metabolism
- Chondroitin Sulfate Proteoglycans/metabolism
- Cicatrix/physiopathology
- Cicatrix/prevention & control
- Glial Fibrillary Acidic Protein/genetics
- Glial Fibrillary Acidic Protein/metabolism
- Graft Survival/physiology
- Growth Cones/metabolism
- Growth Cones/ultrastructure
- Mice
- Mice, Transgenic
- Nerve Fibers, Myelinated/metabolism
- Nerve Fibers, Myelinated/ultrastructure
- Nerve Growth Factor/genetics
- Nerve Growth Factor/metabolism
- Nerve Regeneration/physiology
- Promoter Regions, Genetic/genetics
- Superior Cervical Ganglion/cytology
- Superior Cervical Ganglion/metabolism
- Superior Cervical Ganglion/transplantation
- Sympathetic Fibers, Postganglionic/cytology
- Sympathetic Fibers, Postganglionic/metabolism
- Sympathetic Fibers, Postganglionic/transplantation
- Tissue Transplantation
- Up-Regulation/physiology
Collapse
Affiliation(s)
- David B. Pettigrew
- Department of Neurosurgery, The Neuroscience Institute, University of Cincinnati College of Medicine, ML 0515, Cincinnati, OH 45267-0515, USA
| | - Ya-Qin Li
- Department of Neurosurgery, The Neuroscience Institute, University of Cincinnati College of Medicine, ML 0515, Cincinnati, OH 45267-0515, USA
| | - Charles Kuntz
- Department of Neurosurgery, The Neuroscience Institute, University of Cincinnati College of Medicine, ML 0515, Cincinnati, OH 45267-0515, USA
| | - Keith A. Crutcher
- Department of Neurosurgery, The Neuroscience Institute, University of Cincinnati College of Medicine, ML 0515, Cincinnati, OH 45267-0515, USA
| |
Collapse
|
237
|
Zhu Z, Zhang Q, Yu Z, Zhang L, Tian D, Zhu S, Bu B, Xie M, Wang W. Inhibiting cell cycle progression reduces reactive astrogliosis initiated by scratch injury in vitro and by cerebral ischemia in vivo. Glia 2007; 55:546-58. [PMID: 17243097 DOI: 10.1002/glia.20476] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Astrogliosis occurs in a variety of neuropathological disorders and injuries, and excessive astrogliosis can be devastating to the recovery of neuronal function. In this study, we asked whether reactive astrogliosis can be suppressed in the lesion area by cell cycle inhibition and thus have therapeutic benefits. Reactive astrogliosis induced in either cultured astrocytes by hypoxia or scratch injury, or in a middle cerebral artery occlusion (MCAO) ischemia model were combined to address this issue. In the cultured astrocytes, hypoxia induced a cell cycle activation that was associated with upregulation of the proliferating cell nuclear marker (PCNA). Significantly, the cell cycle inhibitor, olomoucine, inhibited hypoxia-induced cell cycle activation by arresting the cells at G1/S and G2/M in a dose-dependent manner and also reversed hypoxia-induced upregulation of PCNA. Also in the cultured astrocytes, scratch injury induced reactive astrogliosis, such as hypertrophy and an increase in BrdU(+) astrocytes, both of which were ameliorated by olomoucine. In the MCAO ischemia mouse model, dense reactive glial fibrillary acidic protein and PCNA immunoreactivity were evident at the boundary zone of focal cerebral ischemia at days 7 and 30 after MCAO. We found that intraperitoneal olomoucine administration significantly inhibited these astrogliosis-associated changes. To demonstrate further that cell cycle regulation impacts on astrogliosis, cyclin D1 gene knockout mice (cyclin D1(-/-)) were subjected to ischemia, and we found that the percentage of Ki67-positive astrocytes in these mice was markedly reduced in the boundary zone. The number of apoptotic neurons and the lesion volume in cyclin D1(-/-) mice also decreased as compared to cyclin D1(+/+) and cyclin D1(+/-) mice at days 3, 7, and 30 after local cerebral ischemia. Together, these in vitro and in vivo results strongly suggest that astrogliosis can be significantly affected by cell cycle inhibition, which therefore emerges as a promising intervention to attenuate reactive glia-related damage to neuronal function in brain pathology.
Collapse
Affiliation(s)
- Zhou Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Chan CCM, Wong AK, Liu J, Steeves JD, Tetzlaff W. ROCK inhibition with Y27632 activates astrocytes and increases their expression of neurite growth-inhibitory chondroitin sulfate proteoglycans. Glia 2007; 55:369-84. [PMID: 17136770 DOI: 10.1002/glia.20466] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inhibition of Rho-kinase (ROCK) with Y27632 stimulates sprouting by injured corticospinal tract and dorsal column tract axons, and accelerates functional recovery. However, regeneration of these axons across the glial scar was not observed. Here we examined the effects of Y27632 treatment on chondroitin sulfate proteoglycan (CSPG) expression by astrocytes, which are a key component of the reactive gliosis inhibiting axonal regeneration. In vivo, rats underwent a dorsal column transection and were treated with Y27632 via intrathecal pump infusion. Compared with controls, Y27632-treated injury sites displayed exaggerated upregulation of glial fibrillary acid protein and neurocan immunoreactivity along the lesion edge. In vitro, astrocytes assumed a reactive morphology (stellate shape) and increased their expression of CSPGs after Y27632 treatment. Neurite growth by dissociated cortical neurons decreased when cultured on the extracellular matrix (ECM) derived from Y27632-treated astrocytes. This decrease in neurite growth was reversed with chondroitinase-ABC (ChABC) digestion, indicating that the inhibition was due to CSPG depositions within the ECM. Interestingly, conditioned medium (CM) from untreated astrocytes was inhibitory to neurite growth, which was overcome by ChABC digestion. Such inhibitory activity was not found in the CM of Y27632-treated astrocytes. Taken together, these data support a model where ROCK inhibition by Y27632 modifies astrocytic processing of CSPGs, and increases the presence of CSPGs within the ECM while reduces CSPGs in the CM (cerebrospinal fluid in vivo). This increased expression of inhibitory CSPGs in the ECM of the glial scar may counteract the growth promoting effects of ROCK inhibition on axonal growth cones.
Collapse
Affiliation(s)
- Carmen C M Chan
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
239
|
Rossi F, Gianola S, Corvetti L. Regulation of intrinsic neuronal properties for axon growth and regeneration. Prog Neurobiol 2006; 81:1-28. [PMID: 17234322 DOI: 10.1016/j.pneurobio.2006.12.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 11/04/2006] [Accepted: 12/05/2006] [Indexed: 01/29/2023]
Abstract
Regulation of neuritic growth is crucial for neural development, adaptation and repair. The intrinsic growth potential of nerve cells is determined by the activity of specific molecular sets, which sense environmental signals and sustain structural extension of neurites. The expression and function of these molecules are dynamically regulated by multiple mechanisms, which adjust the actual growth properties of each neuron population at different ontogenetic stages or in specific conditions. The neuronal potential for axon elongation and regeneration are restricted at the end of development by the concurrent action of several factors associated with the final maturation of neurons and of the surrounding tissue. In the adult, neuronal growth properties can be significantly modulated by injury, but they are also continuously tuned in everyday life to sustain physiological plasticity. Strict regulation of structural remodelling and neuritic elongation is thought to be required to maintain specific patterns of connectivity in the highly complex mammalian CNS. Accordingly, procedures that neutralize such mechanisms effectively boost axon growth in both intact and injured nervous system. Even in these conditions, however, aberrant connections are only formed in the presence of unusual external stimuli or experience. Therefore, growth regulatory mechanisms play an essentially permissive role by setting the responsiveness of neural circuits to environmental stimuli. The latter exert an instructive action and determine the actual shape of newly formed connections. In the light of this notion, efficient therapeutic interventions in the injured CNS should combine targeted manipulations of growth control mechanisms with task-specific training and rehabilitation paradigms.
Collapse
Affiliation(s)
- Ferdinando Rossi
- Rita Levi Montalcini Centre for Brain Repair, Department of Neuroscience, University of Turin, Corso Raffaello 30, I-10125 Turin, Italy.
| | | | | |
Collapse
|
240
|
Tian DS, Yu ZY, Xie MJ, Bu BT, Witte OW, Wang W. Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal cord injury rat model by the cell cycle inhibitor olomoucine. J Neurosci Res 2006; 84:1053-63. [PMID: 16862564 DOI: 10.1002/jnr.20999] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It is well established that axons of the adult mammalian CNS are capable of regrowing only a limited amount after injury. Astrocytes are believed to play a crucial role in the failure to regenerate, producing multiple inhibitory proteoglycans, such as chondroitin sulphate proteoglycans (CSPGs). After spinal cord injury (SCI), astrocytes become hypertrophic and proliferative and form a dense network of astroglial processes at the site of lesion constituting a physical and biochemical barrier. Down-regulations of astroglial proliferation and inhibitory CSPG production might facilitate axonal regeneration. Recent reports indicated that aberrant activation of cell cycle machinery contributed to overproliferation and apoptosis of cells in various insults. In the present study, we sought to determine whether a cell cycle inhibitior, olomoucine, would decrease neuronal cell death, limit astroglial proliferation and production of inhibitory CSPGs, and eventually enhance the functional compensation after SCI in rats. Our results showed that up-regulations of cell cycle components were closely associated with neuronal cell death and astroglial proliferation as well as the production of CSPGs after SCI. Meanwhile, administration of olomoucine, a selective cell cycle kinase (CDK) inhibitor, has remarkably reduced the up-regulated cell cycle proteins and then decreased neuronal cell death, astroglial proliferation, and accumulation of CSPGs. More importantly, the treatment with olomoucine has also increased expression of growth-associated proteins-43, reduced cavity formation, and improved functional deficits. We consider that suppressing astroglial cell cycle in acute SCIs is beneficial to axonal growth. In the future, therapeutic strategies can be designed to achieve efficient axonal regeneration and functional compensation after traumatic CNS injury.
Collapse
Affiliation(s)
- Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | | | | | | | | |
Collapse
|
241
|
Nitzan A, Kermer P, Shirvan A, Bähr M, Barzilai A, Solomon AS. Examination of cellular and molecular events associated with optic nerve axotomy. Glia 2006; 54:545-56. [PMID: 16906543 DOI: 10.1002/glia.20398] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Analyzing cellular behavior during scar formation and determining the expression of growth inhibiting molecules in the optic nerve and retina following acute optic nerve injury. METHODS A rat model of complete transection of the optic nerve that spares the vascular supply and the neural scaffold was used. The response of the optic nerve and retinas to axotomy was studied by immunological and biochemical approaches. RESULTS Optic nerve axotomy led to massive cell invasion at the site of injury that spread along both sides of the nerve. The cells were microglia, oligodendrocytes, and to a lesser extent astrocytes. A marked induction of semaphorin 3A was evident, especially in the area of the scar, and persisted up to the 28th day of the experiment. Expression of neuropilin-1, a component of the semaphorin 3A receptor, increased following injury. The molecular events associated with axotomy were studied by measuring the levels of semaphorin 3A, p38 MAPK, and ERK1/2 in the retina. Semaphorin 3A levels and the activated form of p38 were elevated 3 days post-axotomy and then declined; ERK1/2 activation levels reached their peak 14 days post axotomy. Acute nerve injury led to morphological alterations in oligodendrocytes, astrocytes, and the extracellular matrix, disrupting the delicate internal organization of the optic nerve. CONCLUSIONS We suggest that cell invasion, semaphorin 3A and neuropilin-1 induction, and disruption of the internal organization of the optic nerve contribute to axotomy-induced degenerative processes.
Collapse
Affiliation(s)
- Anat Nitzan
- Goldschleger Eye Research Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | | | | | | | | | | |
Collapse
|
242
|
Itoh T, Satou T, Nishida S, Hashimoto S, Ito H. Cultured Rat Astrocytes Give Rise to Neural Stem Cells. Neurochem Res 2006; 31:1381-7. [PMID: 17053966 DOI: 10.1007/s11064-006-9186-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Accepted: 09/26/2006] [Indexed: 11/29/2022]
Abstract
Previously, we reported the occurrence of neural stem cells (NSCs) around an area of damage after rat traumatic brain injury (TBI), but it was unclear if this was due to blastgenesis in astrocytes, or to NSCs migrating from the subventricular zone (SVZ). In this study, NSCs were isolated and cultured from cultured type 1 astrocytes taken from newborn rat cortex in which the subventricular zone and hippocampus had been discarded. All cultured type 1 astrocytes showed glial fibrillary acidic protein (GFAP) immunopositivity. Nestin immunopositive spheres were isolated from type 1 astrocytes and cultured in the presence of bFGF and EGF in the medium. Neurospheres differentiated into Tuj1-, GFAP- and A2B5-positive cells after 4 days of culture without bFGF and EGF. These results indicate that isolated neurospheres from brain cortex astrocytes can differentiate into neurons and glia and might contribute to neurogenesis and neuroplasticity.
Collapse
Affiliation(s)
- Tatsuki Itoh
- Department of Pathology, Kinki University School of Medicine, Osaka, Japan.
| | | | | | | | | |
Collapse
|
243
|
Ozawa T, Ohtori S, Inoue G, Aoki Y, Moriya H, Takahashi K. The degenerated lumbar intervertebral disc is innervated primarily by peptide-containing sensory nerve fibers in humans. Spine (Phila Pa 1976) 2006; 31:2418-22. [PMID: 17023849 DOI: 10.1097/01.brs.0000239159.74211.9c] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Immunohistochemical study of the sensory innervation of the human lumbar intervertebral disc. OBJECTIVE To determine the type of sensory fibers innervating human degenerated lumbar intervertebral discs. SUMMARY OF BACKGROUND DATA Sensory neurons involved in pain perception related to inflammation in rats are typically small, peptide-containing neurons immunoreactive for calcitonin gene-related peptide (CGRP). Small non-peptide-containing neurons binding to isolectin B4 (IB4) may also be involved in pain states, such as nerve injury pain. The character of such sensory neurons in humans has not been clarified. METHODS A degenerated, painful lumbar intervertebral disc was harvested from each of 8 patients during surgery. Sections were immunostained for protein gene product 9.5 (PGP 9.5, a general neuronal marker), CGRP, and IB4. The numbers of PGP 9.5- and CGRP-immunoreactive, and IB4-binding nerve fibers in the discs were counted. RESULTS PGP 9.5-immunoreactive fibers were observed in all discs. Nerve fibers immunoreactive for CGRP were also observed in 6 of 8 cases. IB4-binding nerve fibers were not found in any case. CONCLUSIONS Almost all of the nociceptive nerve fibers in the human intervertebral disc are peptide-containing nerve fibers, similar to the rat disc, suggesting that nerve fibers related to inflammation may transmit pain originating from human degenerated intervertebral discs.
Collapse
Affiliation(s)
- Tomoyuki Ozawa
- From the Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | | | | | | | | | | |
Collapse
|
244
|
Zhou FQ, Snider WD. Intracellular control of developmental and regenerative axon growth. Philos Trans R Soc Lond B Biol Sci 2006; 361:1575-92. [PMID: 16939976 PMCID: PMC1664665 DOI: 10.1098/rstb.2006.1882] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 06/19/2006] [Indexed: 01/10/2023] Open
Abstract
Axon growth is a highly regulated process that requires stimulating signals from extracellular factors. The extracellular signals are then transduced to regulate coordinately gene expression and local axon assembly. Growth factors, especially neurotrophins that act via receptor tyrosine kinases, have been heavily studied as extracellular factors that stimulate axon growth. Downstream of receptor tyrosine kinases, recent studies have suggested that phosphatidylinositol-3 kinase (PI3K) regulates local assembly of axonal cytoskeleton, especially microtubules, via glycogen synthase kinase 3beta (GSK-3beta) and multiple microtubule binding proteins. The role of extracellular signal regulated kinase (ERK) signalling in regulation of local axon assembly is less clear, but may involve the regulation of local protein translation. Gene expression during axon growth is regulated by transcription factors, among which cyclic AMP response element binding protein and nuclear factors of activated T-cells (NFATs) are known to be required for neurotrophin (NT)-induced axon extension. In addition to growth factors, extracellular matrix molecules and neuronal activity contribute importantly to control axon growth. Increasingly, evidence suggests that these influences act to enhance growth via coordinating with growth factor signalling. Finally, evidence is emerging that developmental versus regenerative axon growth may be mediated by distinct signalling pathways, both at the level of gene transcription and at the level of local axon assembly.
Collapse
Affiliation(s)
- Feng-Quan Zhou
- Department of Orthopedic Surgery, The Johns Hopkins University School of MedicineBaltimore, MD 21287, USA
- Department of Neuroscience, The Johns Hopkins University School of MedicineBaltimore, MD 21205, USA
| | - William D Snider
- UNC-Neuroscience Center, University of North Carolina at Chapel Hill8109 Neuroscience Research Building, 103 Mason Farm Road, Chapel Hill, NC 27599-7250, USA
| |
Collapse
|
245
|
Liu BP, Cafferty WB, Budel SO, Strittmatter SM. Extracellular regulators of axonal growth in the adult central nervous system. Philos Trans R Soc Lond B Biol Sci 2006; 361:1593-610. [PMID: 16939977 PMCID: PMC1664666 DOI: 10.1098/rstb.2006.1891] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Robust axonal growth is required during development to establish neuronal connectivity. However, stable fibre patterns are necessary to maintain adult mammalian central nervous system (CNS) function. After adult CNS injury, factors that maintain axonal stability limit the recovery of function. Extracellular molecules play an important role in preserving the stability of the adult CNS axons and in restricting recovery from pathological damage. Adult axonal growth inhibitors include a group of proteins on the oligodendrocyte, Nogo-A, myelin-associated glycoprotein, oligodendrocyte-myelin glycoprotein and ephrin-B3, which interact with axonal receptors, such as NgR1 and EphA4. Extracellular proteoglycans containing chondroitin sulphates also inhibit axonal sprouting in the adult CNS, particularly at the sites of astroglial scar formation. Therapeutic perturbations of these extracellular axonal growth inhibitors and their receptors or signalling mechanisms provide a degree of axonal sprouting and regeneration in the adult CNS. After CNS injury, such interventions support a partial return of neurological function.
Collapse
Affiliation(s)
| | | | | | - Stephen M Strittmatter
- Department of Neurology, Yale University School of MedicinePO Box 208018, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
246
|
Yip PK, Wong LF, Pattinson D, Battaglia A, Grist J, Bradbury EJ, Maden M, McMahon SB, Mazarakis ND. Lentiviral vector expressing retinoic acid receptor beta2 promotes recovery of function after corticospinal tract injury in the adult rat spinal cord. Hum Mol Genet 2006; 15:3107-18. [PMID: 16984961 DOI: 10.1093/hmg/ddl251] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Spinal cord injury often results in permanent and devastating neurological deficits and disability. This is due to the limited regenerative capacity of neurones in the central nervous system (CNS). We recently demonstrated that a transcription factor retinoic acid receptor beta2 (RARbeta2) promoted axonal regeneration in adult sensory neurones located peripherally. However, it is not known if RARbeta2 can promote axonal regeneration in cortical neurones of the CNS. Here, we demonstrate that delivery of RARbeta2 via a lentiviral vector to adult dissociated cortical neurones significantly enhances neurite outgrowth on adult cortical cryosections, which normally provide an unfavourable substrate for growth. We also show that lentiviral-mediated transduction of corticospinal neurones resulted in robust transgene expression in layer V corticospinal neurones and their axonal projections in the corticospinal tract (CST) of the spinal cord. Expression of RARbeta2 in these neurones enhanced regeneration of the descending CST fibres after injury to these axons in the mid-cervical spinal cord. Furthermore, we observed functional recovery in sensory and locomotor behavioural tests in RARbeta2-treated animals. These results suggest that a direct and selective delivery of RARbeta2 to the corticospinal neurones promotes long-distance functional regeneration of axons in the spinal cord and may thus offer new therapeutic gene strategy for the treatment of human spinal cord injuries.
Collapse
Affiliation(s)
- Ping K Yip
- Neurorestoration Group, Wolfson CARD, 2 MRC Centre for Developmental Biology, King's College London, Guy's Campus, London Bridge, London SE1 1UL, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Kim BG, Dai HN, Lynskey JV, McAtee M, Bregman BS. Degradation of chondroitin sulfate proteoglycans potentiates transplant-mediated axonal remodeling and functional recovery after spinal cord injury in adult rats. J Comp Neurol 2006; 497:182-98. [PMID: 16705682 PMCID: PMC2570641 DOI: 10.1002/cne.20980] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transplantation of growth-permissive cells or tissues was used to bridge a lesion cavity and induce axonal growth in experimental spinal cord injury (SCI). Axonal interactions between host and transplant may be affected by upregulation of inhibitory chondroitin sulfate proteoglycans (CSPGs) following various transplantation strategies. The extent of axonal growth and functional recovery after transplantation of embryonic spinal cord tissue decreases in adult compared to neonatal host. We hypothesized that CSPGs contribute to the decrease in the extent to which transplant supports axonal remodeling and functional recovery. Expression of CSPGs increased after overhemisection SCI in adult rats but not in neonates. Embryonic spinal cord transplant was surrounded by CSPGs deposited in host cord, and the interface between host and transplant seemed to contain a large amount of CSPGs. Intrathecally delivered chondroitinase ABC (C'ase) improved recovery of distal forelimb usage and skilled motor behavior after C4 overhemisection injury and transplantation in adults. This behavioral recovery was accompanied by an increased amount of raphespinal axons growing into the transplant, and raphespinal innervation to the cervical motor region was promoted by C'ase plus transplant. Moreover, C'ase increased the number of transplanted neurons that grew axons to the host cervical enlargement, suggesting that degradation of CSPGs supports remodeling not only of host axons but also axons from transplanted neurons. Our results suggest that CSPGs constitute an inhibitory barrier to prevent axonal interactions between host and transplant in adults, and degradation of the inhibitory barrier can potentiate transplant-mediated axonal remodeling and functional recovery after SCI.
Collapse
Affiliation(s)
- Byung G Kim
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | | | |
Collapse
|
248
|
Abstract
Damage to the adult CNS often leads to persistent deficits due to the inability of mature axons to regenerate after injury. Mounting evidence suggests that the glial environment of the adult CNS, which includes inhibitory molecules in CNS myelin as well as proteoglycans associated with astroglial scarring, might present a major hurdle for successful axon regeneration. Here, we evaluate the molecular basis of these inhibitory influences and their contributions to the limitation of long-distance axon repair and other types of structural plasticity. Greater insight into glial inhibition is crucial for developing therapies to promote functional recovery after neural injury.
Collapse
Affiliation(s)
- Glenn Yiu
- Division of Neuroscience, Children's Hospital, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
249
|
Heron PM, Sutton BM, Curinga GM, Smith GM, Snow DM. Localized gene expression of axon guidance molecules in neuronal co-cultures. J Neurosci Methods 2006; 159:203-14. [PMID: 16935349 DOI: 10.1016/j.jneumeth.2006.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Revised: 06/30/2006] [Accepted: 07/07/2006] [Indexed: 10/24/2022]
Abstract
Axonal growth cones are guided to their targets by contact-dependent mechanisms or by diffusible chemotropic factors. Axon guidance by these factors typically involves culturing neurons on an acellular substrate which may not represent the in vivo biological environment. We developed two novel in vitro methods to create patterned gene expression of guidance molecules in a physiologically-relevant cellular environment. In the Matrigel assay, a droplet of adenovirus-Matrigel suspension was placed on astrocytes grown in Matrigel. The adenovirus diffused through the gel and transduced underlying astrocytes, creating a radial infection gradient within a localized area. In the second model, recombinant adenovirus was bound to an anti-hexon antibody adsorbed onto stripe patterns of nitrocellulose. Once the cells were added, only those contacting the adenovirus were infected. The outgrowth pattern of chick DRG neurons on NGF, semaphorin 3A and brevican were studied. As expected, results showed robust axonal growth toward NGF as opposed to either secreted Sema 3A or membrane bound brevican, however subtle differences in axonal growth responses were observed in comparison to those obtained with less physiologically-relevant methods. Novel to this technology, the location and area of molecule expression can be controlled and manipulated in an intricate cellular environment.
Collapse
Affiliation(s)
- P M Heron
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | | | | | | | | |
Collapse
|
250
|
Wu D, Shibuya S, Miyamoto O, Itano T, Yamamoto T. Increase of NG2-positive cells associated with radial glia following traumatic spinal cord injury in adult rats. ACTA ACUST UNITED AC 2006; 34:459-69. [PMID: 16902766 DOI: 10.1007/s11068-006-8998-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 09/09/2005] [Accepted: 04/27/2006] [Indexed: 10/24/2022]
Abstract
In the CSN including the spinal cord, NG2 proteoglycan is a marker of oligodendrocyte progenitors. To elucidate the dynamics of the endogenous neural stem (progenitor) cells in adult rats with spinal cord injury (SCI), we examined an immunohistochemical analysis of NG2, GFAP, and 3CB2, a specific marker of radial glia (RG). SD rats were divided into a SCI group (n = 25) and a sham-operated group (n = 5). In the injury group, laminectomy was performed at Th11-12 and contusive compression injury was created by applying a weight of 30 g for 10 min. Rats were sacrificed at 24 h, and 1, 4, 8 and 12 weeks post-injury. Frozen 20-mu m sections of tissue 5 and 10 mm rostral and caudal to the epicenter of injury were prepared. Immunohistochemistry was performed using antibodies against NG2, GFAP and 3CB2. At 4 weeks after injury, NG2-positive glial cells arose from below the pial surface as bipolar cells with processes extending throughout the entire white matter. NG2 expression peaked at 4 weeks after injury, showing a 7-fold increase compared to the 24 h after injury. The NG2-positive cells with processes which increased in the white matter of the spinal cord were GFAP-positive and also co-localized with 3CB2 antigen. The pattern of NG2 expression of these cells was temporally and spatially different from the pattern of NG2 expression that accumulated around the hemorrhagic and necrotic epicenter. These results suggest that NG2 positive cells which derived from subpial layer, may have some lineage to RG after SCI in adult rodents.
Collapse
Affiliation(s)
- Di Wu
- Departments of Orthopaedic Surgery, School of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kagawa, 761-0793, Japan
| | | | | | | | | |
Collapse
|