101
|
Akbik FV, Bhagat SM, Patel PR, Cafferty WBJ, Strittmatter SM. Anatomical plasticity of adult brain is titrated by Nogo Receptor 1. Neuron 2013; 77:859-66. [PMID: 23473316 DOI: 10.1016/j.neuron.2012.12.027] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2012] [Indexed: 11/29/2022]
Abstract
Experience rearranges anatomical connectivity in the brain, but such plasticity is suppressed in adulthood. We examined the turnover of dendritic spines and axonal varicosities in the somatosensory cortex of mice lacking Nogo Receptor 1 (NgR1). Through adolescence, the anatomy and plasticity of ngr1 null mice are indistinguishable from control, but suppression of turnover after age 26 days fails to occur in ngr1-/- mice. Adolescent anatomical plasticity can be restored to 1-year-old mice by conditional deletion of ngr1. Suppression of anatomical dynamics by NgR1 is cell autonomous and is phenocopied by deletion of Nogo-A ligand. Whisker removal deprives the somatosensory cortex of experience-dependent input and reduces dendritic spine turnover in adult ngr1-/- mice to control levels, while an acutely enriched environment increases dendritic spine dynamics in control mice to the level of ngr1-/- mice in a standard environment. Thus, NgR1 determines the low set point for synaptic turnover in adult cerebral cortex.
Collapse
Affiliation(s)
- Feras V Akbik
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neurobiology, Yale University School of Medicine, New Haven, CT 06536, USA
| | | | | | | | | |
Collapse
|
102
|
Ahmed Z, Douglas MR, John G, Berry M, Logan A. AMIGO3 is an NgR1/p75 co-receptor signalling axon growth inhibition in the acute phase of adult central nervous system injury. PLoS One 2013; 8:e61878. [PMID: 23613963 PMCID: PMC3628841 DOI: 10.1371/journal.pone.0061878] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 03/17/2013] [Indexed: 12/18/2022] Open
Abstract
Axon regeneration in the injured adult CNS is reportedly inhibited by myelin-derived inhibitory molecules, after binding to a receptor complex comprised of the Nogo-66 receptor (NgR1) and two transmembrane co-receptors p75/TROY and LINGO-1. However, the post-injury expression pattern for LINGO-1 is inconsistent with its proposed function. We demonstrated that AMIGO3 levels were significantly higher acutely than those of LINGO-1 in dorsal column lesions and reduced in models of dorsal root ganglion neuron (DRGN) axon regeneration. Similarly, AMIGO3 levels were raised in the retina immediately after optic nerve crush, whilst levels were suppressed in regenerating optic nerves, induced by intravitreal peripheral nerve implantation. AMIGO3 interacted functionally with NgR1-p75/TROY in non-neuronal cells and in brain lysates, mediating RhoA activation in response to CNS myelin. Knockdown of AMIGO3 in myelin-inhibited adult primary DRG and retinal cultures promoted disinhibited neurite growth when cells were stimulated with appropriate neurotrophic factors. These findings demonstrate that AMIGO3 substitutes for LINGO-1 in the NgR1-p75/TROY inhibitory signalling complex and suggests that the NgR1-p75/TROY-AMIGO3 receptor complex mediates myelin-induced inhibition of axon growth acutely in the CNS. Thus, antagonizing AMIGO3 rather than LINGO-1 immediately after CNS injury is likely to be a more effective therapeutic strategy for promoting CNS axon regeneration when combined with neurotrophic factor administration.
Collapse
Affiliation(s)
- Zubair Ahmed
- Neuropharmacology and Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom.
| | | | | | | | | |
Collapse
|
103
|
Yan Y, Li T, Liu L, Zhou H. Effect of tonifying liver and kidney-essence herbs on expression of Nogo-A and p75(NTR) in cerebral ischemic stroke rats model. J TRADIT CHIN MED 2013; 32:664-8. [PMID: 23427407 DOI: 10.1016/s0254-6272(13)60089-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To observe the effect of tonifying liver and kidney-essence herbs on expression of a nerve regeneration inhibitor, Nogo for neuron A (Nogo-A), and its associated signaling molecule, low-affinity neurotrophin receptor p75 (p75(NTR)), in rats with cerebral ischemic stroke (CIS), with the aim of exploring the possible mechanism of tonifying liver and kidney-essence herbs in recovery following injury to the central nervous system. METHODS A cerebral ischemic stroke model in SD rats was established with the suture-occlusion method. Successful model rats were divided into placebo and herb groups at random; sham-operated and control groups were set up simultaneously. Each of these groups was divided into six subgroups at random. Expression of Nogo-A and p75(NTR) was evaluated with immunofluorescence microscopy at days 3, and weeks 1, 2, 3, 4 and 8 after administration. RESULTS Tonifying liver and kidney-essence herbs suppressed the expression of Nogo-A and p75(NTR) (P < 0.05 and P < 0.01, respectively). CONCLUSION Suppressing the expression of Nogo-A and p75(NTR) is possibly one of the mechanisms underlying the ability of tonifying liver and kidney-essence herbs to promote recovery of the injured central nervous system.
Collapse
Affiliation(s)
- Yongmei Yan
- Department of Encephalopathy, Shaanxi University of Traditional Medicine Hospital, Shaanxi 712000, China
| | | | | | | |
Collapse
|
104
|
VanGuilder Starkey HD, Sonntag WE, Freeman WM. Increased hippocampal NgR1 signaling machinery in aged rats with deficits of spatial cognition. Eur J Neurosci 2013; 37:1643-58. [PMID: 23438185 DOI: 10.1111/ejn.12165] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 01/18/2013] [Accepted: 01/21/2013] [Indexed: 01/31/2023]
Abstract
Myelin-associated inhibitor/NgR1 signaling has important roles in modulation of synaptic plasticity, with demonstrated effects on cognitive function. We have previously demonstrated that NgR1 and its ligands are upregulated in the hippocampus of aged rats with impaired spatial learning and memory, but it is unknown whether increased expression of these proteins indicates a potential increase in pathway signaling because NgR1 requires co-receptors for signal transduction through RhoA. Two co-receptor complexes have been identified to date, comprised of NgR1 and LINGO-1, and either p75 or TROY. In this study, we assessed the expression of LINGO-1, p75 and TROY, and the downstream effector RhoA in mature adult (12 months) and aged (26 months) male Fischer 344/Brown Norway hybrid rats classified as cognitively impaired or cognitively intact by Morris water maze testing. The hippocampal distribution of NgR1 and its co-receptors was assessed to determine whether receptor/co-receptor interaction, and therefore signaling through this pathway, is possible. Protein expression of LINGO-1, p75, TROY and RhoA was significantly elevated in cognitively impaired, but not intact, aged rats compared with mature adults, and expression levels correlated significantly with water maze performance. Co-localization of NgR1 with LINGO-1, p75 and TROY was observed in hippocampal neurons of aged, cognitively impaired rats. Further, expression profiles of NgR1 pathway components were demonstrated to classify rats as cognitively intact or cognitively impaired with high accuracy. Together, this suggests that hippocampal induction of this pathway is a conserved phenomenon in cognitive decline that may impair learning and memory by suppressing neuronal plasticity.
Collapse
Affiliation(s)
- Heather D VanGuilder Starkey
- Department of Pharmacology, R130 Hershey Center for Applied Research, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | |
Collapse
|
105
|
Falavigna A, da Costa JC. Mesenchymal autologous stem cells. World Neurosurg 2013; 83:236-50. [PMID: 23402865 DOI: 10.1016/j.wneu.2013.02.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 01/24/2013] [Accepted: 02/05/2013] [Indexed: 02/07/2023]
Abstract
The use of cell-based therapies for spinal cord injuries has recently gained prominence as a potential therapy or component of a combination strategy. Experimental and clinical studies have been performed using mesenchymal stem cell therapy to treat spinal cord injuries with encouraging results. However, there have been reports on the adverse effects of these stem cell-based therapies, especially in the context of tumor modulation. This article surveys the literature relevant to the potential of mesenchymal autologous stem cells for spinal cord injuries and their clinical implications.
Collapse
Affiliation(s)
- Asdrubal Falavigna
- Department of Neurosurgery, Medical School of the University of Caxias do Sul, Caxias do Sul, Brazil.
| | - Jaderson Costa da Costa
- Neurology Service and Instituto do Cérebro, Pontifical Catholic University of Rio Grande do Sul, Brazil
| |
Collapse
|
106
|
Fan Q, Bohannon KP, Longnecker R. Drosophila Schneider 2 (S2) cells: a novel tool for studying HSV-induced membrane fusion. Virology 2013; 437:100-9. [PMID: 23399037 DOI: 10.1016/j.virol.2013.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/31/2012] [Accepted: 01/04/2013] [Indexed: 12/17/2022]
Abstract
Drosophila S2 cells and mammalian CHO-K1 cells were used to investigate the requirements for HSV-1 cell fusion. Infection assays indicated S2 cells were not permissive for HSV-1. HVEM and nectin-1 mediated cell fusion between CHO-K1 cells and S2 cells when either CHO-K1 or S2 cells were used as target cells. Interestingly, PILRα did not mediate fusion between CHO-K1 or S2 cells due to a glycosylation defect of PILRα and gB in S2 cells. Fusion activity was not detected for any receptor tested when S2 cells were used both as target cells and effector cells indicating S2 cells may lack a key cellular factor present in mammalian cells that is required for cell fusion. Thus, insect cells may provide a novel tool to study the interaction of HSV-1 glycoproteins and cellular factors required for fusion, as well as a means to identify unknown cellular factors required for HSV replication.
Collapse
Affiliation(s)
- Qing Fan
- Northwestern University, Department of Microbiology-Immunology, Ward 6-241, 303 East Chicago Avenue, Chicago, IL 60611, United States
| | | | | |
Collapse
|
107
|
Tong J, Liu W, Wang X, Han X, Hyrien O, Samadani U, Smith DH, Huang JH. Inhibition of Nogo-66 receptor 1 enhances recovery of cognitive function after traumatic brain injury in mice. J Neurotrauma 2013; 30:247-58. [PMID: 22967270 DOI: 10.1089/neu.2012.2493] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Central nervous system (CNS) axons recover poorly following injury because of the expression of myelin-derived inhibitors of axonal outgrowth such as Nogo, myelin-associated glycoprotein (MAG), and oligodendrocyte-myelin glycoprotein (OMgp), all of which bind to the Nogo-66 receptor 1 (NgR1). Herein we examine the role of NgR1 in the recovery of motor and cognitive function after traumatic brain injury (TBI) using a controlled cortical impact (CCI) model in NgR1 knockout (KO) and wild-type (WT) mice. Four weeks post-injury, scores on the Novel Object Recognition test were significantly increased in NgR1 KO mice compared with WT mice (p<0.05), but motor behavior test scores did not differ significantly between the two groups. Nissl staining showed that NgR1 KO mice had less brain injury volume 2 weeks after CCI (p<0.05). Histological analysis revealed more doublecortin (DCX+) cells (p<0.01) and more Ki-67+ cells in the contralateral dentate gyrus (DG) (p<0.05) 2 weeks after CCI in NgR1 KO mice than in WT. Furthermore, DCX+ cells still retained their longer processes in KO mice (p<0.01) 4 weeks following trauma. The number of bromodeoxyuridine (BrdU)+ cells did not differ between the two groups at 4 weeks post-trauma, but KO mice had higher numbers of cells that co-stained with NeuN, a marker of mature neurons. Increased transcription of growth-associated protein (GAP)-43 in both the injured and contralateral sides of the hippocampus (both p<0.05) was detected in NgR1 KO mice relative to WT. These data suggest that NgR1 negatively influences plasticity and cognitive recovery after TBI.
Collapse
Affiliation(s)
- Jing Tong
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | |
Collapse
|
108
|
The promotion of functional recovery and nerve regeneration after spinal cord injury by lentiviral vectors encoding Lingo-1 shRNA delivered by Pluronic F-127. Biomaterials 2012; 34:1686-700. [PMID: 23211450 DOI: 10.1016/j.biomaterials.2012.11.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 11/10/2012] [Indexed: 12/11/2022]
Abstract
Lingo-1 is selectively expressed on both oligodendrocytes and neurons in the central nervous system (CNS) and serves as a key negative regulator of nerve regeneration, implying a therapeutic target for spinal cord injury (SCI). Here we described a strategy to knock-down Lingo-1 expression in vivo using lentiviral vectors encoding Lingo-1 short harpin interfering RNA (shRNA) delivered by Pluronic F-127 (PF-127) gel, a non-cytotoxic scaffold and gene delivery carrier, after the complete transection of the T10 spinal cord in adult rats. We showed administration of PF-127 encapsulating Lingo-1 shRNA lentiviral vectors efficiently down-regulated the expression of Lingo-1, and exhibited transduction efficiency comparable to using vectors alone in oligodendrocyte culture in vitro. Furthermore, similar silencing effects and higher transfection efficiency were observed in vivo when Lingo-1 shRNA was co-delivered to the injured site by PF-127 gel with lower viral concentrations. Cografting of gel and Lingo-1 RNAi significantly promoted functional recovery and nerve regeneration, enhanced neurite outgrowth and synapses formation, preserved myelinated axons, and induced the proliferation of glial cells. In addition, the combined implantation also improved neuronal survival and inhibited cell apoptosis, which may be associated with the attenuation of endoplasmic reticulum (ER) stress after SCI. Together, our data indicated that delivering Lingo-1 shRNA by gel scaffold was a valuable treatment approach to SCI and PF-127 delivery of viral vectors to the spinal cord may provide strategy to study and develop therapies for SCI.
Collapse
|
109
|
Yang J, Han Y, Ye W, Liu F, Zhuang K, Wu G. Alpha tocopherol treatment reduces the expression of Nogo-A and NgR in rat brain after traumatic brain injury. J Surg Res 2012. [PMID: 23207171 DOI: 10.1016/j.jss.2012.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Neurite outgrowth inhibitor-A (Nogo-A), myelin-associated glycoprotein, and oligodendrocyte myelin glycoprotein are three myelin-associated proteins that act as inhibitors to central nervous system regeneration. Neurite outgrowth inhibitor-A imposes the strongest effect on inhibiting axonal regeneration after traumatic brain injury. Alpha-tocopherol, a member of the vitamin E family, is recognized as an active antioxidative substance. Its use has not been well studied in brain injury research, especially in axonal regeneration research. METHODS We obtained 99 intact adult male Sprague-Dawley rats (200-250 g) from the Experimental Animal Center of Central South University. We used the modified method of Freeney to generate moderate brain injury in the rats. We injected 600 mg/kg α-tocopherol intraperitoneally daily as traumatic brain injury (TBI) treatment. Then, we performed behavioral tests in the corresponding time point, examined brain tissues after hematoxylin-eosin staining to identify changes in cell morphology, and performed immunohistochemical staining and quantitative real-time polymerase chain reaction to detect the expression of NoGo and Nogo receptor (NgR) in brain tissue. RESULTS For the Neurological Severity Scores of rats, there were obvious differences among the three groups at the corresponding time points. Standard hematoxylin-eosin staining showed that the brain structure of a sham-operated group of rats was clear, uniform, and compact. A TBI group exhibited hemorrhage, edema, inflammatory cell infiltration, condensed nuclei, and necrosis. We also saw glial cells and fibrous tissue proliferation. The α-tocopherol-treated TBI group had similar but less severe changes than the TBI group. Expression of Nogo-A and NgR increased after TBI compared with the sham-operated group. However, Nogo-A and NgR expression was significantly lower in the α-tocopherol-treated TBI group compared with the TBI group. Similarly, results showed that functional neurological deficits among rats in the α-tocopherol-treated TBI group were less pronounced than in the TBI group (model group). CONCLUSIONS Our data demonstrate that α-tocopherol-treated rats had reduced microscopic evidence of brain damage. Alpha-tocopherol reduced Nogo-A and NgR expression in brain tissue after traumatic brain injury and promoted nerve regeneration. Alpha-tocopherol treatment of TBI rats had a neuroprotective role in their recovery.
Collapse
Affiliation(s)
- Jinfu Yang
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | | | | | | | | | | |
Collapse
|
110
|
Hutson TH, Foster E, Dawes JM, Hindges R, Yáñez-Muñoz RJ, Moon LDF. Lentiviral vectors encoding short hairpin RNAs efficiently transduce and knockdown LINGO-1 but induce an interferon response and cytotoxicity in central nervous system neurones. J Gene Med 2012; 14:299-315. [PMID: 22499506 DOI: 10.1002/jgm.2626] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Knocking down neuronal LINGO-1 using short hairpin RNAs (shRNAs) might enhance axon regeneration in the central nervous system (CNS). Integration-deficient lentiviral vectors have great potential as a therapeutic delivery system for CNS injuries. However, recent studies have revealed that shRNAs can induce an interferon response resulting in off-target effects and cytotoxicity. METHODS CNS neurones were transduced with integration-deficient lentiviral vectors in vitro. The transcriptional effect of shRNA expression was analysed using quantitative real time-polymerase chain reaction and northern blots were used to assess shRNA production. RESULTS Integration-deficient lentiviral vectors efficiently transduced CNS neurones and knocked down LINGO-1 mRNA in vitro. However, an increase in cell death was observed when lentiviral vectors encoding an shRNA were applied or when high vector concentrations were used. We demonstrate that high doses of vector or the use of vectors encoding shRNAs can induce an up-regulation of interferon-stimulated genes (2',5'-oligoadenylate synthase 1 and protein kinase R although not myxovirus resistance 1) and a down-regulation of off-target genes (including p75(NTR) and Nogo receptor 1). Furthermore, the northern blot demonstrated that these negative consequences occur even when lentiviral vectors express low levels of shRNAs. Taken together, these results may explain why neurite outgrowth was not enhanced on an inhibitory substrate following transduction with lentiviral vectors encoding an shRNA targeting LINGO-1. CONCLUSIONS These findings highlight the importance of including appropriate controls to verify silencing specificity and the requirement to check for an interferon response when conducting RNA interference experiments. However, the potential benefits that RNA interference and viral vectors offer to gene-based therapies to CNS injuries cannot be overlooked and demand further investigation.
Collapse
Affiliation(s)
- Thomas H Hutson
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK. thomas.hutson@kcl. ac.uk
| | | | | | | | | | | |
Collapse
|
111
|
The potential for cellular therapy combined with growth factors in spinal cord injury. Stem Cells Int 2012; 2012:826754. [PMID: 23091499 PMCID: PMC3471462 DOI: 10.1155/2012/826754] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 08/19/2012] [Accepted: 08/28/2012] [Indexed: 12/18/2022] Open
Abstract
Any traumatic spinal cord injury (SCI) may cause symptoms ranging from pain to complete loss of motor and sensory functions below the level of the injury. Currently, there are over 2 million SCI patients worldwide. The cost of their necessary continuing care creates a burden for the patient, their families, and society. Presently, few SCI treatments are available and none have facilitated neural regeneration and/or significant functional improvement. Research is being conducted in the following areas: pathophysiology, cellular therapies (Schwann cells, embryonic stem cells, induced pluripotent stem cells, mesenchymal stem cells, olfactory ensheathing cells), growth factors (BDNF), inhibitory molecules (NG2, myelin protein), and combination therapies (cell grafts and neurotrophins, cotransplantation). Results are often limited because of the inhibitory environment created following the injury and the limited regenerative potential of the central nervous system. Therapies that show promise in small animal models may not transfer to nonhuman primates and humans. None of the research has resulted in remarkable improvement, but many areas show promise. Studies have suggested that a combination of therapies may enhance results and may be more effective than a single therapy. This paper reviews and discusses the most promising new SCI research including combination therapies.
Collapse
|
112
|
Sharma K, Selzer ME, Li S. Scar-mediated inhibition and CSPG receptors in the CNS. Exp Neurol 2012; 237:370-8. [PMID: 22836147 PMCID: PMC5454774 DOI: 10.1016/j.expneurol.2012.07.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 07/14/2012] [Indexed: 11/21/2022]
Abstract
Severed axons in adult mammals do not regenerate appreciably after central nervous system (CNS) injury due to developmentally determined reductions in neuron-intrinsic growth capacity and extracellular environment for axon elongation. Chondroitin sulfate proteoglycans (CSPGs), which are generated by reactive scar tissues, are particularly potent contributors to the growth-limiting environment in mature CNS. Thus, surmounting the strong inhibition by CSPG-rich scar is an important therapeutic goal for achieving functional recovery after CNS injuries. As of now, the main in vivo approach to overcoming inhibition by CSPGs is enzymatic digestion with locally applied chondroitinase ABC (ChABC), but several disadvantages may prevent using this bacterial enzyme as a therapeutic option for patients. A better understanding of the molecular mechanisms underlying CSPG action is needed in order to develop more effective therapies to overcome CSPG-mediated inhibition of axon regeneration and/or sprouting. Because of their large size and dense negative charges, CSPGs were thought to act by non-specifically hindering the binding of matrix molecules to their cell surface receptors through steric interactions. Although this may be true, recent studies indicate that two members of the leukocyte common antigen related (LAR) phosphatase subfamily, protein tyrosine phosphatase σ (PTPσ) and LAR, are functional receptors that bind CSPGs with high affinity and mediate CSPG inhibitory effects. CSPGs also may act by binding to two receptors for myelin-associated growth inhibitors, Nogo receptors 1 and 3 (NgR1 and NgR3). If confirmed, it would suggest that CSPGs have multiple mechanisms by which they inhibit axon growth, making them especially potent and difficult therapeutic targets. Identification of CSPG receptors is not only important for understanding the scar-mediated growth suppression, but also for developing novel and selective therapies to promote axon sprouting and/or regeneration after CNS injuries, including spinal cord injury (SCI).
Collapse
Affiliation(s)
- Kartavya Sharma
- Department of Neurology and Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, Texas 75390-8813, USA
| | - Michael E. Selzer
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Shuxin Li
- Department of Neurology and Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, Texas 75390-8813, USA
| |
Collapse
|
113
|
Lagier-Tourenne C, Polymenidou M, Hutt KR, Vu AQ, Baughn M, Huelga SC, Clutario KM, Ling SC, Liang TY, Mazur C, Wancewicz E, Kim AS, Watt A, Freier S, Hicks GG, Donohue JP, Shiue L, Bennett CF, Ravits J, Cleveland DW, Yeo GW. Divergent roles of ALS-linked proteins FUS/TLS and TDP-43 intersect in processing long pre-mRNAs. Nat Neurosci 2012; 15:1488-97. [PMID: 23023293 DOI: 10.1038/nn.3230] [Citation(s) in RCA: 578] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 09/06/2012] [Indexed: 02/06/2023]
Abstract
FUS/TLS (fused in sarcoma/translocated in liposarcoma) and TDP-43 are integrally involved in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. We found that FUS/TLS binds to RNAs from >5,500 genes in mouse and human brain, primarily through a GUGGU-binding motif. We identified a sawtooth-like binding pattern, consistent with co-transcriptional deposition of FUS/TLS. Depletion of FUS/TLS from the adult nervous system altered the levels or splicing of >950 mRNAs, most of which are distinct from RNAs dependent on TDP-43. Abundance of only 45 RNAs was reduced after depletion of either TDP-43 or FUS/TLS from mouse brain, but among these were mRNAs that were transcribed from genes with exceptionally long introns and that encode proteins that are essential for neuronal integrity. Expression levels of a subset of these were lowered after TDP-43 or FUS/TLS depletion in stem cell-derived human neurons and in TDP-43 aggregate-containing motor neurons in sporadic ALS, supporting a common loss-of-function pathway as one component underlying motor neuron death from misregulation of TDP-43 or FUS/TLS.
Collapse
Affiliation(s)
- Clotilde Lagier-Tourenne
- Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Mishra M, Lee S, Lin MK, Yamashita T, Heese K. Characterizing the neurite outgrowth inhibitory effect of Mani. FEBS Lett 2012; 586:3018-23. [DOI: 10.1016/j.febslet.2012.06.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 06/14/2012] [Accepted: 06/18/2012] [Indexed: 12/01/2022]
|
115
|
Guo X, Zahir T, Mothe A, Shoichet MS, Morshead CM, Katayama Y, Tator CH. The Effect of Growth Factors and Soluble Nogo-66 Receptor Protein on Transplanted Neural Stem/Progenitor Survival and Axonal Regeneration after Complete Transection of Rat Spinal Cord. Cell Transplant 2012; 21:1177-97. [DOI: 10.3727/096368911x612503] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adult central mammalian axons show minimal regeneration after spinal cord injury due to loss of oligodendrocytes, demyelination of surviving axons, absence of growth-promoting molecules, and inhibitors of axonal outgrowth. In the present study, we attempted to address these impediments to regeneration by using a combinatory strategy to enhance cell survival and regeneration after complete spinal cord transection (SCT) in adult rats. The strategy comprised: 1) adult rat brain-derived neural stem/progenitor cells (NSPCs) preseeded on laminin-coated chitosan channels; 2) extramedullary chitosan channels to promote axonal regrowth and reduce the barrier caused by scarring; 3) local delivery of a novel rat soluble Nogo-66 receptor protein [NgR(310)ecto-Fc, referred to as NgR] to block the inhibitory effect of myelin-based inhibitors; and 4) local delivery of basic fibroblast growth factor, epidermal growth factor, and platelet-derived growth factor to enhance survival and promote differentiation of transplanted cells. Compared with our previous studies where brain-derived NSPCs preseeded in extramedullary chitosan channels were implanted in the same SCT model but without growth factors and NgR, the present channel–growth factor combination produced greater numbers of surviving NSPCs after SCT. Also, the growth factors promoted preferential differentiation of NSPCs toward oligodendrocytes, while NgR significantly decreased astrocytic differentiation of NSPCs. NgR alone or in combination with NSPCs significantly enhanced the total number of myelinated fibers in the bridge and increased the area of the bridging tissue between the cord stumps. The combination of NgR, growth factors, and NSPCs had synergistic effect on bridge formation. However, only a small number of descending corticospinal tract axons grew into the central portions of the bridges as shown by anterograde tracing of the corticospinal tract with BDA. The majority of the regenerated axons in the channels originated from local host neurons adjacent to the tissue bridges. In conclusion, we showed that growth factors increased survival of transplanted NSPCs whereas NgR enhanced axonal regeneration, but the combination did not have additive effects on functional recovery or regeneration.
Collapse
Affiliation(s)
- Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Tasneem Zahir
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Mothe
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Molly S. Shoichet
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Cindi M. Morshead
- Department of Surgery and Institute of Medical Sciences, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Yusuke Katayama
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Charles H. Tator
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
116
|
Lee JK, Zheng B. Role of myelin-associated inhibitors in axonal repair after spinal cord injury. Exp Neurol 2012; 235:33-42. [PMID: 21596039 PMCID: PMC3170678 DOI: 10.1016/j.expneurol.2011.05.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Revised: 04/19/2011] [Accepted: 05/01/2011] [Indexed: 12/15/2022]
Abstract
Myelin-associated inhibitors of axon growth, including Nogo, MAG and OMgp, have been the subject of intense research. A myriad of experimental approaches have been applied to investigate the potential of targeting these molecules to promote axonal repair after spinal cord injury. However, there are still conflicting results on their role in axon regeneration and therefore a lack of a cohesive mechanism on how these molecules can be targeted to promote axon repair. One major reason may be the lack of a clear definition of axon regeneration in the first place. Nevertheless, recent data from genetic studies in mice indicate that the roles of these molecules in CNS axon repair may be more intricate than previously envisioned.
Collapse
Affiliation(s)
- Jae K Lee
- University of California San Diego, Department of Neurosciences, 9500 Gilman Drive, MC 0691, La Jolla, CA 92093-0691, USA.
| | | |
Collapse
|
117
|
Akbik F, Cafferty WBJ, Strittmatter SM. Myelin associated inhibitors: a link between injury-induced and experience-dependent plasticity. Exp Neurol 2012; 235:43-52. [PMID: 21699896 PMCID: PMC3189418 DOI: 10.1016/j.expneurol.2011.06.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 04/26/2011] [Accepted: 06/07/2011] [Indexed: 01/01/2023]
Abstract
In the adult, both neurologic recovery and anatomical growth after a CNS injury are limited. Two classes of growth inhibitors, myelin associated inhibitors (MAIs) and extracellular matrix associated inhibitors, limit both functional recovery and anatomical rearrangements in animal models of spinal cord injury. Here we focus on how MAIs limit a wide spectrum of growth that includes regeneration, sprouting, and plasticity in both the intact and lesioned CNS. Three classic myelin associated inhibitors, Nogo-A, MAG, and OMgp, signal through their common receptors, Nogo-66 Receptor-1 (NgR1) and Paired-Immunoglobulin-like-Receptor-B (PirB), to regulate cytoskeletal dynamics and inhibit growth. Initially described as inhibitors of axonal regeneration, subsequent work has demonstrated that MAIs also limit activity and experience-dependent plasticity in the intact, adult CNS. MAIs therefore represent a point of convergence for plasticity that limits anatomical rearrangements regardless of the inciting stimulus, blurring the distinction between injury studies and more "basic" plasticity studies.
Collapse
Affiliation(s)
- Feras Akbik
- Cellular Neuroscience, Neurodegeneration and Repair Program, and Departments of Neurology and of Neurobiology, Yale School of Medicine, New Haven, CT USA
| | - William B. J. Cafferty
- Cellular Neuroscience, Neurodegeneration and Repair Program, and Departments of Neurology and of Neurobiology, Yale School of Medicine, New Haven, CT USA
| | - Stephen M. Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, and Departments of Neurology and of Neurobiology, Yale School of Medicine, New Haven, CT USA
| |
Collapse
|
118
|
Liang Y, Liu Z, Shuai X, Wang W, Liu J, Bi W, Wang C, Jing X, Liu Y, Tao E. Delivery of cationic polymer-siRNA nanoparticles for gene therapies in neural regeneration. Biochem Biophys Res Commun 2012; 421:690-5. [DOI: 10.1016/j.bbrc.2012.03.155] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 03/21/2012] [Indexed: 11/30/2022]
|
119
|
Wang T, Xiong JQ, Ren XB, Sun W. The role of Nogo-A in neuroregeneration: A review. Brain Res Bull 2012; 87:499-503. [DOI: 10.1016/j.brainresbull.2012.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 02/21/2012] [Accepted: 02/24/2012] [Indexed: 01/24/2023]
|
120
|
He J, Wang XM, Spector M, Cui FZ. Scaffolds for central nervous system tissue engineering. FRONTIERS OF MATERIALS SCIENCE 2012; 6:1-25. [DOI: 10.1007/s11706-012-0157-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
121
|
Kopp MA, Liebscher T, Niedeggen A, Laufer S, Brommer B, Jungehulsing GJ, Strittmatter SM, Dirnagl U, Schwab JM. Small-molecule-induced Rho-inhibition: NSAIDs after spinal cord injury. Cell Tissue Res 2012; 349:119-32. [PMID: 22350947 DOI: 10.1007/s00441-012-1334-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 01/16/2012] [Indexed: 01/16/2023]
Abstract
Limited axonal plasticity within the central nervous system (CNS) is a major restriction for functional recovery after CNS injury. The small GTPase RhoA is a key molecule of the converging downstream cascade that leads to the inhibition of axonal re-growth. The Rho-pathway integrates growth inhibitory signals derived from extracellular cues, such as chondroitin sulfate proteoglycans, Nogo-A, myelin-associated glycoprotein, oligodendrocyte-myelin glycoprotein, Ephrins and repulsive guidance molecule-A, into the damaged axon. Consequently, the activation of RhoA results in growth cone collapse and finally outgrowth failure. In turn, the inhibition of RhoA-activation blinds the injured axon to its growth inhibitory environment resulting in enhanced axonal sprouting and plasticity. This has been demonstrated in various CNS-injury models for direct RhoA-inhibition and for downstream/upstream blockade of the RhoA-associated pathway. In addition, RhoA-inhibition reduces apoptotic cell death and secondary damage and improves locomotor recovery in clinically relevant models after experimental spinal cord injury (SCI). Unexpectedly, a subset of "small molecules" from the group of non-steroid anti-inflammatory drugs, particularly the FDA-approved ibuprofen, has recently been identified as (1) inhibiting RhoA-activation, (2) enhancing axonal sprouting/regeneration, (3) protecting "tissue at risk" (neuroprotection) and (4) improving motor recovery confined to realistic therapeutical time-frames in clinically relevant SCI models. Here, we survey the effect of small-molecule-induced RhoA-inhibition on axonal plasticity and neurofunctional outcome in CNS injury paradigms. Furthermore, we discuss the body of preclinical evidence for a possible clinical translation with a focus on ibuprofen and illustrate putative risks and benefits for the treatment of acute SCI.
Collapse
Affiliation(s)
- M A Kopp
- Department of Neurology and Experimental Neurology, Spinal Cord Injury Research, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Herpes B virus utilizes human nectin-1 but not HVEM or PILRα for cell-cell fusion and virus entry. J Virol 2012; 86:4468-76. [PMID: 22345445 DOI: 10.1128/jvi.00041-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To investigate the requirements of herpesvirus entry and fusion, the four homologous glycoproteins necessary for herpes simplex virus (HSV) fusion were cloned from herpes B virus (BV) (or macacine herpesvirus 1, previously known as cercopithecine herpesvirus 1) and cercopithecine herpesvirus 2 (CeHV-2), both related simian simplexviruses belonging to the alphaherpesvirus subfamily. Western blots and cell-based enzyme-linked immunosorbent assay (ELISA) showed that glycoproteins gB, gD, and gH/gL were expressed in whole-cell lysates and on the cell surface. Cell-cell fusion assays indicated that nectin-1, an HSV-1 gD receptor, mediated fusion of cells expressing glycoproteins from both BV and CeHV-2. However, herpesvirus entry mediator (HVEM), another HSV-1 gD receptor, did not facilitate BV- and CeHV-2-induced cell-cell fusion. Paired immunoglobulin-like type 2 receptor alpha (PILRα), an HSV-1 gB fusion receptor, did not mediate fusion of cells expressing glycoproteins from either simian virus. Productive infection with BV was possible only with nectin-1-expressing cells, indicating that nectin-1 mediated entry while HVEM and PILRα did not function as entry receptors. These results indicate that these alphaherpesviruses have differing preferences for entry receptors. The usage of the HSV-1 gD receptor nectin-1 may explain interspecies transfer of the viruses, and altered receptor usage may result in altered virulence, tropism, or pathogenesis in the new host. A heterotypic cell fusion assay resulting in productive fusion may provide insight into interactions that occur to trigger fusion. These findings may be of therapeutic significance for control of deadly BV infections.
Collapse
|
123
|
The Nogo-66 receptor family in the intact and diseased CNS. Cell Tissue Res 2012; 349:105-17. [PMID: 22311207 DOI: 10.1007/s00441-012-1332-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 01/16/2012] [Indexed: 10/14/2022]
Abstract
The Nogo-66 receptor family (NgR) consists in three glycophosphatidylinositol (GPI)-anchored receptors (NgR1, NgR2 and NgR3), which are primarily expressed by neurons in the central and peripheral mammalian nervous system. NgR1 was identified as serving as a high affinity binding protein for the three classical myelin-associated inhibitors (MAIs) Nogo-A, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp), which limit axon regeneration and sprouting in the injured brain. Recent studies suggest that NgR signaling may also play an essential role in the intact adult CNS in restricting axonal and synaptic plasticity and are involved in neurodegenerative diseases, particularly in Alzheimer's disease pathology through modulation of β-secretase cleavage. Here, we outline the biochemical properties of NgRs and their functional roles in the intact and diseased CNS.
Collapse
|
124
|
Expression and function of myelin-associated proteins and their common receptor NgR on oligodendrocyte progenitor cells. Brain Res 2012; 1437:1-15. [DOI: 10.1016/j.brainres.2011.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 12/01/2011] [Accepted: 12/03/2011] [Indexed: 11/30/2022]
|
125
|
Chiu H, Alqadah A, Chuang CF, Chang C. C. elegans as a genetic model to identify novel cellular and molecular mechanisms underlying nervous system regeneration. Cell Adh Migr 2012; 5:387-94. [PMID: 21975547 DOI: 10.4161/cam.5.5.17985] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Research into conditions that improve axon regeneration has the potential to open a new door for treatment of brain injury caused by stroke and neurodegenerative diseases of aging, such as Alzheimer, by harnessing intrinsic neuronal ability to reorganize itself. Elucidating the molecular mechanisms of axon regeneration should shed light on how this process becomes restricted in the postnatal stage and in CNS and therefore could provide therapeutic targets for developing strategy to improve axon regeneration in adult CNS. In this review, we first discuss the general view about nerve regeneration and the advantages of using C. elegans as a model system to study axon regeneration. We then compare the conserved regeneration patterns and molecular mechanisms between C. elegans and vertebrates. Lastly, we discuss the power of femtosecond laser technology and its application in axon regeneration research.
Collapse
Affiliation(s)
- Hui Chiu
- Division of Developmental Biology; Cincinnati Children's Hospital Research Foundation; Cincinnati, OH, USA
| | | | | | | |
Collapse
|
126
|
Kern F, Sarg B, Stasyk T, Hess D, Lindner H. The Nogo receptor 2 is a novel substrate of Fbs1. Biochem Biophys Res Commun 2012; 417:977-81. [PMID: 22206664 PMCID: PMC3269754 DOI: 10.1016/j.bbrc.2011.12.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 12/13/2011] [Indexed: 11/15/2022]
Abstract
Members of the Nogo66 receptor family (NgR) are closely associated with nerve growth inhibition and plasticity in the CNS. All three members, NgR1, NgR2 and NgR3, are GPI anchored and highly glycosylated proteins. The binding and signaling properties of NgR1 are well described, but largely unknown for NgR2. At present the only known ligands are myelin associated glycoprotein (MAG) and amyloid beta precursor protein (APP). Despite the requirement of co-receptors for signaling no other binding partner has been uncovered. To learn more about the interactome of NgR2 we performed pull down experiments and were able to identify F-box protein that recognizes sugar chain 1 (Fbs1) as binding partner. We confirmed this finding with co-immunoprecipitations and in vitro binding assays and showed that the binding is mediated by the substrate recognition domain of Fbs1. As a substrate recognition protein of the SCF complex, Fbs1 binding leads to polyubiquitination and finally degradation of its substrates. This is the first time a member of the Nogo receptor family has been connected with an intracellular degradation pathway, which has not only implications for its production, but also for amyloid deposition in Alzheimer’s disease.
Collapse
Affiliation(s)
- Florian Kern
- Neurobiochemistry - Biocenter, Innsbruck Medical University, Austria.
| | | | | | | | | |
Collapse
|
127
|
Abstract
Spinal cord injury (SCI) has multiple consequences, ranging from molecular imbalances to glial scar formation to functional impairments. It is logical to think that a combination of single treatments implemented in the right order and at the right time will be required to repair the spinal cord. However, the single treatments that compose the combination therapy will need to be chosen with caution as many have multiple outcomes that may or may not be synergistic. Single treatments may also elicit unwanted side-effects and/or effects that would decrease the repair potential of other components and/or the entire combination therapy. In this chapter a number of single treatments are discussed with respect to their multiplicity of action. These include strategies to boost growth and survival (such as neurotrophins and cyclic AMP) and strategies to reduce inhibitory factors (such as antimyelin-associated growth inhibitors and digestion of glial scar-associated inhibitors). We also present an overview of combination therapies that have successfully or unsuccessfully been tested in the laboratory using animal models. To effectively design a combination therapy a number of considerations need to be made such as the nature and timing of the treatments and the method for delivery. This chapter discusses these issues as well as considerations related to chronic SCI and the logistics of bringing combination therapies to the clinic.
Collapse
Affiliation(s)
- M Oudega
- Departments of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
128
|
Abstract
AbstractCentral nervous system (CNS) injuries affect all levels of society indiscriminately, resulting in functional and behavioral deficits with devastating impacts on life expectancies, physical and emotional wellbeing. Considerable literature exists describing the pathophysiology of CNS injuries as well as the cellular and molecular factors that inhibit regrowth and regeneration of damaged connections. Based on these data, numerous therapeutic strategies targeting the various factors of repair inhibition have been proposed and on-going assessment has demonstrated some promising results in the laboratory environ. However, several of these treatment strategies have subsequently been taken into clinical trials but demonstrated little to no improvement in patient outcomes. As a result, options for clinical interventions following CNS injuries remain limited and effective restorative treatment strategies do not as yet exist. This review discusses some of the current animal models, with focus on nonhuman primates, which are currently being modeled in the laboratory for the study of CNS injuries. Last, we review the current understanding of the mechanisms underlying repair/regrowth inhibition and the current trends in experimental treatment strategies that are being assessed for potential translation to clinical applications.
Collapse
|
129
|
Luo X, Park KK. Neuron-Intrinsic Inhibitors of Axon Regeneration. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012. [DOI: 10.1016/b978-0-12-398309-1.00008-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
130
|
Skaper SD. Neuronal growth-promoting and inhibitory cues in neuroprotection and neuroregeneration. Methods Mol Biol 2012; 846:13-22. [PMID: 22367797 DOI: 10.1007/978-1-61779-536-7_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
During development of the nervous system, neurons extend axons over considerable distances in a highly stereospecific fashion in order to innervate their targets in an appropriate manner. This involves the recognition, by the axonal growth cone, of guidance cues that determine the pathway taken by the axons. These guidance cues can act to promote and/or repel growth cone advance. The directed growth of axons is partly governed by cell adhesion molecules (CAMs) on the neuronal growth cone that bind to CAMs on the surface of other axons or nonneuronal cells. In vitro assays have established the importance of the CAMs ((neural cell adhesion molecule NCAM), N-cadherin, and L1) in promoting axonal growth over cells. Compelling evidence implicates the fibroblast growth factor receptor tyrosine kinase as the primary signal transduction molecule in the CAM pathway. CAMs are important constituents of synapses, and they appear to play important and diverse roles in regulating synaptic plasticity associated with learning and memory. Synthetic NCAM peptide mimetics corresponding to the binding site of NCAM for the fibroblast growth factor receptor promote synaptogenesis, enhance presynaptic function, and facilitate memory consolidation. Dimeric versions of functional binding motifs of N-cadherin behave as N-cadherin agonists, promoting both neuritogenesis and neuronal cell survival. Negative extracellular signals that physically direct neurite growth have also been described. The latter include the myelin inhibitory proteins, Nogo, myelin-associated glycoprotein, and oligodendrocyte-myelin glycoprotein. Potentiation of outgrowth-promoting signals, together with antagonism of myelin proteins or their convergent receptor, NgR, and its second messenger pathways, may provide new opportunities in the rational design of treatments for acute brain injury and neurodegenerative disorders.
Collapse
Affiliation(s)
- Stephen D Skaper
- Department of Pharmacology and Anesthesiology, University of Padova, Padova, Italy.
| |
Collapse
|
131
|
|
132
|
Fawcett JW, Schwab ME, Montani L, Brazda N, Müller HW. Defeating inhibition of regeneration by scar and myelin components. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:503-22. [PMID: 23098733 DOI: 10.1016/b978-0-444-52137-8.00031-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Axon regeneration and the sprouting processes that underlie plasticity are blocked by inhibitory factors in the central nervous system (CNS) environment, several of which are upregulated after injury. The major inhibitory molecules are those associated with myelin and those associated with the glial scar. In myelin, NogoA, MAG, and OMgp are present on normal oligodendrocytes and on myelin debris. They act partly via the Nogo receptor, partly via an unidentified amino-Nogo receptor. In the glial scar, chondroitin sulphate proteoglycans, semaphorins, and the formation of a collagen-based membrane are all inhibitory. Methods to counteract these forms of inhibition have been identified, and these treatments promote axon regeneration in the damaged spinal cord, and in some cases recovery of function through enhanced plasticity.
Collapse
Affiliation(s)
- James W Fawcett
- Cambridge University Centre for Brain Repair, Cambridge, UK.
| | | | | | | | | |
Collapse
|
133
|
Steinbach K, McDonald CL, Reindl M, Schweigreiter R, Bandtlow C, Martin R. Nogo-receptors NgR1 and NgR2 do not mediate regulation of CD4 T helper responses and CNS repair in experimental autoimmune encephalomyelitis. PLoS One 2011; 6:e26341. [PMID: 22096481 PMCID: PMC3214013 DOI: 10.1371/journal.pone.0026341] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/25/2011] [Indexed: 11/28/2022] Open
Abstract
Myelin-associated inhibition of axonal regrowth after injury is considered one important factor that contributes to regeneration failure in the adult central nervous system (CNS). Blocking strategies targeting this pathway have been successfully applied in several nerve injury models, including experimental autoimmune encephalomyelitis (EAE), suggesting myelin-associated inhibitors (MAIs) and functionally related molecules as targets to enhance regeneration in multiple sclerosis. NgR1 and NgR2 were identified as interaction partners for the myelin proteins Nogo-A, MAG and OMgp and are probably mediating their growth-inhibitory effects on axons, although the in vivo relevance of this pathway is currently under debate. Recently, alternative functions of MAIs and NgRs in the regulation of immune cell migration and T cell differentiation have been described. Whether and to what extent NgR1 and NgR2 are contributing to Nogo and MAG-related inhibition of neuroregeneration or immunomodulation during EAE is currently unknown. Here we show that genetic deletion of both receptors does not promote functional recovery during EAE and that NgR1 and NgR2-mediated signals play a minor role in the development of CNS inflammation. Induction of EAE in Ngr1/2-double mutant mice resulted in indifferent disease course and tissue damage when compared to WT controls. Further, the development of encephalitogenic CD4+ Th1 and Th17 responses was unchanged. However, we observed a slightly increased leukocyte infiltration into the CNS in the absence of NgR1 and NgR2, indicating that NgRs might be involved in the regulation of immune cell migration in the CNS. Our study demonstrates the urgent need for a more detailed knowledge on the multifunctional roles of ligands and receptors involved in CNS regeneration failure.
Collapse
Affiliation(s)
- Karin Steinbach
- Institute for Neuroimmunology and Clinical MS-Research, Hamburg, Germany
| | - Claire L. McDonald
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | | | - Christine Bandtlow
- Department of Neurobiochemistry, Innsbruck Medical University, Innsbruck, Austria
| | - Roland Martin
- Institute for Neuroimmunology and Clinical MS-Research, Hamburg, Germany
- * E-mail:
| |
Collapse
|
134
|
Gerin CG, Madueke IC, Perkins T, Hill S, Smith K, Haley B, Allen SA, Garcia RP, Paunesku T, Woloschak G. Combination strategies for repair, plasticity, and regeneration using regulation of gene expression during the chronic phase after spinal cord injury. Synapse 2011; 65:1255-81. [DOI: 10.1002/syn.20903] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
135
|
Arocho LC, Figueroa JD, Torrado AI, Santiago JM, Vera AE, Miranda JD. Expression profile and role of EphrinA1 ligand after spinal cord injury. Cell Mol Neurobiol 2011; 31:1057-69. [PMID: 21603973 PMCID: PMC3216482 DOI: 10.1007/s10571-011-9705-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 04/30/2011] [Indexed: 10/18/2022]
Abstract
Spinal cord injury (SCI) triggers the re-expression of inhibitory molecules present in early stages of development, contributing to prevention of axonal regeneration. Upregulation of EphA receptor tyrosine kinases after injury suggest their involvement in the nervous system's response to damage. However, the expression profile of their ephrinA ligands after SCI is unclear. In this study, we determined the expression of ephrinA ligands after contusive SCI. Adult Sprague-Dawley female rats were injured using the MASCIS impactor device at the T10 vertebrae, and levels of ephrinA mRNA and protein determined at different time points. Identification of the cell phenotype expressing the ephrin ligand and colocalization with Eph receptors was performed with immunohistochemistry and confocal microscopy. Behavioral studies were made, after blocking ephrinA1 expression with antisense (AS) oligonucleotides, to assess hindlimb locomotor activity. Real-time PCR demonstrated basal mRNA levels of ephrin (A1, A2, A3, and A5) in the adult spinal cord. Interestingly, ephrinA1 was the only ligand whose mRNA levels were significantly altered after SCI. Although ephrinA1 mRNA levels increased after 2 weeks and remain elevated, we did not observe this pattern at the protein level as revealed by western blot analysis. Immunohistochemical studies showed ephrinA1 expression in reactive astrocytes, axons, and neurons and also their colocalization with EphA4 and A7 receptors. Behavioral studies revealed worsening of locomotor activity when ephrinA1 expression was reduced. This study suggests that ephrinA1 ligands play a role in the pathophysiology of SCI.
Collapse
Affiliation(s)
- Luz C. Arocho
- Physiology Department, University of Puerto Rico School of Medicine, P.O. Box 365067, San Juan, PR 00936-5067 USA
| | - Johnny D. Figueroa
- Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA 92354 USA
| | - Aranza I. Torrado
- Physiology Department, University of Puerto Rico School of Medicine, P.O. Box 365067, San Juan, PR 00936-5067 USA
| | - José M. Santiago
- Physiology Department, University of Puerto Rico School of Medicine, P.O. Box 365067, San Juan, PR 00936-5067 USA
| | - Ariel E. Vera
- Physiology Department, University of Puerto Rico School of Medicine, P.O. Box 365067, San Juan, PR 00936-5067 USA
| | - Jorge D. Miranda
- Physiology Department, University of Puerto Rico School of Medicine, P.O. Box 365067, San Juan, PR 00936-5067 USA
| |
Collapse
|
136
|
Current and future therapeutic strategies for functional repair of spinal cord injury. Pharmacol Ther 2011; 132:57-71. [DOI: 10.1016/j.pharmthera.2011.05.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 05/09/2011] [Indexed: 12/26/2022]
|
137
|
Cho Y, Borgens RB. Polymer and nano-technology applications for repair and reconstruction of the central nervous system. Exp Neurol 2011; 233:126-44. [PMID: 21985867 DOI: 10.1016/j.expneurol.2011.09.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 09/16/2011] [Accepted: 09/26/2011] [Indexed: 01/17/2023]
Abstract
The hydrophilic polymer PEG and its related derivatives, have served as therapeutic agents to reconstruct the phospholipid bilayers of damaged cell membranes by erasing defects in the plasmalemma. The special attributes of hydrophilic polymers when in contact with cell membranes have been used for several decades since these well-known properties have been exploited in the manufacture of monoclonal antibodies. However, while traditional therapeutic efforts to combat traumatic injuries of the central nervous system (CNS) have not been successful, nanotechnology-based drug delivery has become a new emerging strategy with the additional promise of targeted membrane repair. As such, this potential use of nanotechnology provides new avenues for nanomedicine that uses nanoparticles themselves as the therapeutic agent in addition to their other functionalities. Here we will specifically address new advances in experimental treatment of Spinal Cord and Traumatic Brain injury (SCI and TBI respectively). We focus on the concept of repair of the neurolemma and axolemma in the acute stage of injury, with less emphasis on the worthwhile, and voluminous, issues concerning regenerative medicine/nanomedicine. It is not that the two are mutually exclusive - they are not. However, the survival of the neuron and the tissues of white matter are critical to any further success in what will likely be a multi-component therapy for TBI and SCI. This review includes a brief explanation of the characteristics of traumatic spinal cord injury SCI, the biological basis of the injuries, and the treatment opportunities of current polymer-based therapies. In particular, we update our own progress in such applications for CNS injuries with various suggestions and discussion, primarily nanocarrier-based drug delivery systems. The application of nanoparticles as drug-delivery vehicles to the CNS may likely be advantageous over existing molecular-based therapies. As a "proof-of-concept", we will discuss the recent investigations that have preferentially facilitated repair and functional recovery from breaches in neural membranes via rapid sealing and reassembly of the compromised site with silica or chitosan nanoparticles.
Collapse
Affiliation(s)
- Youngnam Cho
- Center for Paralysis Research, School of Veterinary Medicine, Purdue University, W. Lafayette, IN 47907, USA
| | | |
Collapse
|
138
|
McDonald CL, Steinbach K, Kern F, Schweigreiter R, Martin R, Bandtlow CE, Reindl M. Nogo receptor is involved in the adhesion of dendritic cells to myelin. J Neuroinflammation 2011; 8:113. [PMID: 21906273 PMCID: PMC3177896 DOI: 10.1186/1742-2094-8-113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/09/2011] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Nogo-66 receptor NgR1 and its structural homologue NgR2 are binding proteins for a number of myelin-associated inhibitory factors. After neuronal injury, these inhibitory factors are responsible for preventing axonal outgrowth via their interactions with NgR1 and NgR2 expressed on neurons. In vitro, cells expressing NgR1/2 are inhibited from adhering to and spreading on a myelin substrate. Neuronal injury also results in the presence of dendritic cells (DCs) in the central nervous system, where they can come into contact with myelin debris. The exact mechanisms of interaction of immune cells with CNS myelin are, however, poorly understood. METHODS Human DCs were differentiated from peripheral blood monocytes and mouse DCs were differentiated from wild type and NgR1/NgR2 double knockout bone marrow precursors. NgR1 and NgR2 expression were determined with quantitative real time PCR and immunoblot, and adhesion of cells to myelin was quantified. RESULTS We demonstrate that human immature myeloid DCs express NgR1 and NgR2, which are then down-regulated upon maturation. Human mature DCs also adhere to a much higher extent to a myelin substrate than immature DCs. We observe the same effect when the cells are plated on Nogo-66-His (binding peptide for NgR1), but not on control proteins. Mature DCs taken from Ngr1/2 knockout mice adhere to a much higher extent to myelin compared to wild type mouse DCs. In addition, Ngr1/2 knockout had no effect on in vitro DC differentiation or phenotype. CONCLUSIONS These results indicate that a lack of NgR1/2 expression promotes the adhesion of DCs to myelin. This interaction could be important in neuroinflammatory disorders such as multiple sclerosis in which peripheral immune cells come into contact with myelin debris.
Collapse
Affiliation(s)
- Claire L McDonald
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
139
|
Twohig JP, Cuff SM, Yong AA, Wang ECY. The role of tumor necrosis factor receptor superfamily members in mammalian brain development, function and homeostasis. Rev Neurosci 2011; 22:509-33. [PMID: 21861782 DOI: 10.1515/rns.2011.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tumor necrosis factor receptor superfamily (TNFRSF) members were initially identified as immunological mediators, and are still commonly perceived as immunological molecules. However, our understanding of the diversity of TNFRSF members' roles in mammalian physiology has grown significantly since the first discovery of TNFRp55 (TNFRSF1) in 1975. In particular, the last decade has provided evidence for important roles in brain development, function and the emergent field of neuronal homeostasis. Recent evidence suggests that TNFRSF members are expressed in an overlapping regulated pattern during neuronal development, participating in the regulation of neuronal expansion, growth, differentiation and regional pattern development. This review examines evidence for non-immunological roles of TNFRSF members in brain development, function and maintenance under normal physiological conditions. In addition, several aspects of brain function during inflammation will also be described, when illuminating and relevant to the non-immunological role of TNFRSF members. Finally, key questions in the field will be outlined.
Collapse
Affiliation(s)
- Jason P Twohig
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, Wales, UK
| | | | | | | |
Collapse
|
140
|
Sato Y, Iketani M, Kurihara Y, Yamaguchi M, Yamashita N, Nakamura F, Arie Y, Kawasaki T, Hirata T, Abe T, Kiyonari H, Strittmatter SM, Goshima Y, Takei K. Cartilage acidic protein-1B (LOTUS), an endogenous Nogo receptor antagonist for axon tract formation. Science 2011; 333:769-73. [PMID: 21817055 DOI: 10.1126/science.1204144] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Neural circuitry formation depends on the molecular control of axonal projection during development. By screening with fluorophore-assisted light inactivation in the developing mouse brain, we identified cartilage acidic protein-1B as a key molecule for lateral olfactory tract (LOT) formation and named it LOT usher substance (LOTUS). We further identified Nogo receptor-1 (NgR1) as a LOTUS-binding protein. NgR1 is a receptor of myelin-derived axon growth inhibitors, such as Nogo, which prevent neural regeneration in the adult. LOTUS suppressed Nogo-NgR1 binding and Nogo-induced growth cone collapse. A defasciculated LOT was present in lotus-deficient mice but not in mice lacking both lotus- and ngr1. These findings suggest that endogenous antagonism of NgR1 by LOTUS is crucial for normal LOT formation.
Collapse
Affiliation(s)
- Yasufumi Sato
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Wang YT, Lu XM, Zhu F, Huang P, Yu Y, Zeng L, Long ZY, Wu YM. The use of a gold nanoparticle-based adjuvant to improve the therapeutic efficacy of hNgR-Fc protein immunization in spinal cord-injured rats. Biomaterials 2011; 32:7988-98. [PMID: 21784510 DOI: 10.1016/j.biomaterials.2011.07.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 07/04/2011] [Indexed: 10/18/2022]
Abstract
As a common receptor for three myelin associated inhibitors, Nogo-66 receptor (NgR) mediates their inhibitory activities on neurite outgrowth in the adult mammalian central nervous system (CNS). Therapeutic vaccination protocol targeting NgR emulsified with Freund's adjuvant (FA) has been used in spinal cord injury (SCI) models. However, the vaccine emulsified with FA may induce some side effects, which are not suitable for further clinical application. As an adjuvant, gold nanoparticles (GNPs) could stimulate a stronger immune response without producing detectable toxicity and physiological damage than FA. There is, however, uncertainty regarding the efficacy of axon regeneration and neuroprotection in vaccines with GNPs as an adjuvant. In this investigation, a recombinant protein vaccine targeting NgR, human NgR-Fc (hNgR-Fc) fusion protein conjugated with 15 nm GNPs was prepared and its effects on axonal regeneration and functional recovery in spinal cord-injured rats were investigated. The results showed that adult rats immunized with the protein vaccine produced higher titers of anti-NgR antibody than that with FA, and the antisera promoted neurite outgrowth in presence of MAG in vitro. In a spinal cord dorsal hemisection model, vaccine immunized with GNPs promoted axonal regeneration more effectively than FA, resulted in significant protection from neuronal loss, and improved functional recovery. Thus, as an adjuvant, 15 nm GNPs can effectively boost the immunogenicity of hNgR-Fc protein vaccine, and promote the repair of spinal cord-injured rats. The utilization of GNPs, for clinical considerations, may be a more beneficial supplement than FA to the promising therapeutic vaccination strategy for promoting SCI repair.
Collapse
Affiliation(s)
- Yong-Tang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Matsushita H, Endo S, Kobayashi E, Sakamoto Y, Kobayashi K, Kitaguchi K, Kuroki K, Söderhäll A, Maenaka K, Nakamura A, Strittmatter SM, Takai T. Differential but competitive binding of Nogo protein and class i major histocompatibility complex (MHCI) to the PIR-B ectodomain provides an inhibition of cells. J Biol Chem 2011; 286:25739-47. [PMID: 21636572 PMCID: PMC3138294 DOI: 10.1074/jbc.m110.157859] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 05/23/2011] [Indexed: 11/06/2022] Open
Abstract
Binding of class I MHC molecules (MHCI) to an inhibitory receptor, PIR-B, expressed on B cells and myeloid cells provides constitutive cellular inhibition, thus ensuring peripheral tolerance. Recent unexpected findings pointed to a novel inhibitory role of PIR-B in neurite regeneration through binding to three axonal outgrowth inhibitors of myelin, including Nogo. Thus, it becomes interesting to determine whether the actions of the inhibitory myelin proteins and MHCI could coexist independently or be mutually exclusive as to the PIR-B-mediated immune and neural cell inhibition. Here, we present data supporting the competition of Nogo- and MHCI-mediated inhibition where they coexist. Kinetic analyses of Nogo and MHCI binding to the whole or a part of the recombinant PIR-B ectodomain revealed that PIR-B binds with higher affinity to Nogo than MHCI and that the MHCI binding only occurred with the N-terminal domains of PIR-B, whereas Nogo binding occurred with either the N- or C-terminal ectodomains. Importantly, kinetic tests indicated that the binding to PIR-B of Nogo and MHCI was competitive. Both endogenous and exogenous Nogo intensified the PIR-B-mediated suppression of interleukin-6 release from lipopolysaccharide-stimulated wild-type, but not PIR-B-deficient, cultured mast cells, indicating that PIR-B mediates Nogo-induced inhibition. Thus, we propose a novel mechanism by which PIR-B-mediated regulation is achieved differentially but competitively via MHCI and Nogo in cells of the immune system.
Collapse
Affiliation(s)
- Haruka Matsushita
- From the Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Shota Endo
- From the Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Eiji Kobayashi
- From the Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Yuzuru Sakamoto
- From the Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
- the Center for General Education, Division of Human Sciences, Tohoku Institute of Technology, Sendai 982-8577, Japan
| | - Keisuke Kobayashi
- From the Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Kohji Kitaguchi
- From the Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Kimiko Kuroki
- the Division of Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Arvid Söderhäll
- Sidec Technologies AB, Kista, Stockholm SE-164 40, Sweden, and
| | - Katsumi Maenaka
- the Division of Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Akira Nakamura
- From the Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Stephen M. Strittmatter
- the Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06536-0812
| | - Toshiyuki Takai
- From the Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
143
|
Semavina M, Saha N, Kolev MV, Goldgur Y, Giger RJ, Himanen JP, Nikolov DB. Crystal structure of the Nogo-receptor-2. Protein Sci 2011; 20:684-9. [PMID: 21308849 DOI: 10.1002/pro.597] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The inhibition of axon regeneration upon mechanical injury is dependent on interactions between Nogo receptors (NgRs) and their myelin-derived ligands. NgRs are composed of a leucine-rich repeat (LRR) region, thought to be structurally similar among the different isoforms of the receptor, and a divergent "stalk" region. It has been shown by others that the LRR and stalk regions of NgR1 and NgR2 have distinct roles in conferring binding affinity to the myelin associated glycoprotein (MAG) in vivo. Here, we show that purified recombinant full length NgR1 and NgR2 maintain significantly higher binding affinity for purified MAG as compared to the isolated LRR region of either NgR1 or NgR2. We also present the crystal structure of the LRR and part of the stalk regions of NgR2 and compare it to the previously reported NgR1 structure with respect to the distinct signaling properties of the two receptor isoforms.
Collapse
Affiliation(s)
- Mariya Semavina
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York 10065, USA
| | | | | | | | | | | | | |
Collapse
|
144
|
Ueno M, Yamashita T. Strategies for regenerating injured axons after spinal cord injury - insights from brain development. Biologics 2011; 2:253-64. [PMID: 19707358 PMCID: PMC2721354 DOI: 10.2147/btt.s2715] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Axonal regeneration does not occur easily after an adult central nervous system (CNS) injury. Various attempts have partially succeeded in promoting axonal regeneration after the spinal cord injury (SCI). Interestingly, several recent therapeutic concepts have emerged from or been tightly linked to the researches on brain development. In a developing brain, remarkable and dynamic axonal elongation and sprouting occur even after the injury; this finding is essential to the development of a therapy for SCI. In this review, we overview the revealed mechanism of axonal tract formation and plasticity in the developing brain and compare the differences between a developing brain and a lesion site in an adult brain. One of the differences is that mature glial cells participate in the repair process in the case of adult injuries. Interestingly, these cells express inhibitory molecules that impede axonal regeneration such as myelin-associated proteins and the repulsive guidance molecules found originally in the developing brain for navigating axons to specific routes. Some reports have clearly elucidated that any treatment designed to suppress these inhibitory cues is beneficial for promoting regeneration and plasticity after an injury. Thus, understanding the developmental process will provide us with an important clue for designing therapeutic strategies for recovery from SCI.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita-shi, Osaka 565-0871, Japan
| | | |
Collapse
|
145
|
de Wit J, Hong W, Luo L, Ghosh A. Role of leucine-rich repeat proteins in the development and function of neural circuits. Annu Rev Cell Dev Biol 2011; 27:697-729. [PMID: 21740233 DOI: 10.1146/annurev-cellbio-092910-154111] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The nervous system consists of an ensemble of billions of neurons interconnected in a highly specific pattern that allows proper propagation and integration of neural activities. The organization of these specific connections emerges from sequential developmental events including axon guidance, target selection, and synapse formation. These events critically rely on cell-cell recognition and communication mediated by cell-surface ligands and receptors. Recent studies have uncovered central roles for leucine-rich repeat (LRR) domain-containing proteins, not only in organizing neural connectivity from axon guidance to target selection to synapse formation, but also in various nervous system disorders. Their versatile LRR domains, in particular, serve as key sites for interactions with a wide diversity of binding partners. Here, we focus on a few exquisite examples of secreted or membrane-associated LRR proteins in Drosophila and mammals and review the mechanisms by which they regulate diverse aspects of nervous system development and function.
Collapse
Affiliation(s)
- Joris de Wit
- Neurobiology Section, Division of Biology, University of California, San Diego, La Jolla, California 92093-0366, USA
| | | | | | | |
Collapse
|
146
|
Zhao X, Wu J, Kuang F, Wang J, Ju G. Silencing of Nogo-A in rat oligodendrocyte cultures enhances process branching. Neurosci Lett 2011; 499:32-6. [PMID: 21624429 DOI: 10.1016/j.neulet.2011.05.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 05/13/2011] [Indexed: 01/25/2023]
Abstract
The myelin-associated protein Nogo-A is a well-known inhibitor for axonal regeneration and compensatory plasticity, yet functions of endogenous Nogo-A in oligodendrocyte differentiation are not as clear. As oligodendrocyte matures, its processes branch and eventually form lamellae that ensheath target axons. The present study examined the effects of decreased levels of Nogo-A on the development of oligodendrocytes. The siRNA mediated Nogo-A silencing in these cells did not change their proliferation rates identified by BrdU incorporation assay and neither the expression of stage specific oligodendrocyte makers as identified by qRT-PCR and immunostaining. But knockdown the expression of Nogo-A significantly enhances the process branching complexity by Sholl analysis. Current results suggest a novel role for Nogo-A in maintaining a restricted branching phenotype in oligodendrocytes process outgrowth, which is a key step towards myelin membrane sheet formation and myelination.
Collapse
Affiliation(s)
- Xianghui Zhao
- The Fourth Military Medical University, Institute of Neuroscience, Shaanxi, Xi'an 710032, PR China.
| | | | | | | | | |
Collapse
|
147
|
Huebner EA, Kim BG, Duffy PJ, Brown RH, Strittmatter SM. A multi-domain fragment of Nogo-A protein is a potent inhibitor of cortical axon regeneration via Nogo receptor 1. J Biol Chem 2011; 286:18026-36. [PMID: 21454605 PMCID: PMC3093876 DOI: 10.1074/jbc.m110.208108] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 03/09/2011] [Indexed: 11/06/2022] Open
Abstract
Nogo-A limits axon regeneration and functional recovery after central nervous system injury in adult mammals. Three regions of Nogo-A (Nogo-A-24, Nogo-66, and Nogo-C39) interact with the neuronal Nogo-66 receptor 1 (NgR1). Nogo-66 also interacts with a structurally unrelated cell surface receptor, paired immunoglobulin-like receptor (PirB). We show here that the other two NgR1-interacting domains, Nogo-A-24 and Nogo-C39, also bind to PirB with high affinity. A purified 22-kDa protein containing all three NgR1- and PirB-interacting domains (Nogo-22) is a substantially more potent growth cone-collapsing molecule than Nogo-66 for chick dorsal root ganglion neurons and mature cortical neurons. Moreover, Nogo-22 inhibits axon regeneration of mature cortical neurons in vitro more potently than does Nogo-66. Although all three NgR1-interacting domains of Nogo-A also interact with PirB, expression of PirB in mature cortical cultures is nearly undetectable. Consistent with a relatively minor role for PirB in mature cortical neurons, Nogo-22 inhibition of axon regeneration is abolished by genetic deletion of NgR1. Thus, NgR1 is the predominant receptor for Nogo-22 in regenerating cortical neurons.
Collapse
Affiliation(s)
- Eric A. Huebner
- From the Department of Neurology and Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, Connecticut 06536 and
| | - Byung G. Kim
- From the Department of Neurology and Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, Connecticut 06536 and
- the Brain Disease Research Center, Institute for Medical Sciences, and Department of Neurology, Ajou University School of Medicine, Suwon 443-749, Korea
| | - Philip J. Duffy
- From the Department of Neurology and Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, Connecticut 06536 and
| | - Rebecca H. Brown
- From the Department of Neurology and Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, Connecticut 06536 and
| | - Stephen M. Strittmatter
- From the Department of Neurology and Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, Connecticut 06536 and
| |
Collapse
|
148
|
Starkey ML, Schwab ME. Anti-Nogo-A and training: can one plus one equal three? Exp Neurol 2011; 235:53-61. [PMID: 21530508 DOI: 10.1016/j.expneurol.2011.04.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 03/15/2011] [Accepted: 04/07/2011] [Indexed: 12/14/2022]
Abstract
Following spinal cord injury (SCI) the adult central nervous system (CNS) has a limited but substantial capacity for repair and plastic reorganisation. The degree of reorganisation is determined by a number of factors such as the extent and location of the lesion, the remaining circuit activity within the CNS and the age at injury. However, even in the best cases this spontaneous reorganisation does not lead to full recovery of the affected behaviour but instead often results in a functionally successful but compensatory strategy. Current SCI research focuses on enhancing fibre tract (re-)growth and recovery processes. Two currently promising approaches are the neutralisation of CNS growth inhibitory factors, and rehabilitative training of remaining networks. Independently, both approaches can lead to substantial functional recovery and anatomical reorganisation. In this review we focus on Nogo-A, a neurite growth inhibitory protein present in the adult CNS, and its role in regenerative and plastic growth following SCI. We then discuss the efforts of rehabilitative training and the potential combination of the two therapies.
Collapse
Affiliation(s)
- Michelle L Starkey
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| | | |
Collapse
|
149
|
Peng Y, Zhang QL, Xu D, Wang YP, Qin XY. Small hairpin RNA interference of the Nogo receptor inhibits oxygen-glucose deprivation-induced damage in rat hippocampal slice cultures. Neuropathology 2011; 30:565-73. [PMID: 20337950 DOI: 10.1111/j.1440-1789.2010.01102.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In adult mammals, CNS damage does not repair well spontaneously. The Nogo receptor (NgR) signaling pathway prevents axonal regrowth and promotes neuronal apoptosis. This pathway, and pathways like it, may be part of the reason why nerves do not regrow. A number of preclinical experiments inhibiting portions of the NgR pathway have yielded limited induction of nerve repair. Here, we developed a small hairpin RNA (shRNA) to knock down NgR expression. With the use of rat hippocampal slices in tissue culture, we induced neuronal damage similar to that of ischemia-reperfusion injury by exposing the cultured tissues to oxygen-glucose deprivation. We then assayed the effect of NgR knockdown in this model system. Adenovirally delivered NgR shRNA decreased NgR mRNA and protein expression. Thirty minutes of oxygen-glucose deprivation resulted in widespread tissue damage, including apoptosis and loss of neurite extension, 72 h after termination of oxygen-glucose deprivation. The NgR shRNA knockdown reduced, but did not eliminate, the effects of oxygen-glucose deprivation. Thus, NgR shRNA shows promise as a potential tool for the treatment of nerve damage.
Collapse
Affiliation(s)
- Yan Peng
- Laboratory of Stem Cell and Tissue Engineering and First Affiliated Hospital, Chongqing Medical University, Yuanjiagang, Chongqing City, China
| | | | | | | | | |
Collapse
|
150
|
Jeon EJ, Xu N, Xu L, Hansen MR. Influence of central glia on spiral ganglion neuron neurite growth. Neuroscience 2011; 177:321-34. [PMID: 21241783 PMCID: PMC3057386 DOI: 10.1016/j.neuroscience.2011.01.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 01/05/2011] [Accepted: 01/06/2011] [Indexed: 10/18/2022]
Abstract
Spiral ganglion neurons (SGNs) extend processes that interact with Schwann cells (SCs) and with oligodendrocytes (OLs) and astrocytes (ACs). We investigated the ability of these glial cells to support SGN neurite growth. In the presence of cultured ACs, OLs and SCs, SGN neurites tended to follow SCs and OLs and cross-over ACs. Most neurites initially followed the type of glial cell on which the neuronal cell body was found. To determine the influence of homogeneous populations of glia on neurite growth, SG explants were plated on cultured SCs, ACs or OLs. The number of neurites/explant extending onto SCs (463.89±16.25) was significantly greater than the number extending onto ACs (111.38±38.73) or OLs (6.75±2.21), indicating that populations of central glia inhibit SGN neurite growth. Treatment with cell-permeant cpt-cAMP or forskolin (FSK) each significantly increased the number of neurites on OLs (133.54±25.59 and 292.25±83.57, respectively). cpt-cAMP and FSK each also increased the number of neurites on ACs (213.19±36.06 and 208.64±59.25, respectively), however the difference was not significant compared with control. The neurites on ACs and OLs failed to grow radially in a well-fasciculated pattern as on SCs. In explants plated on the borders of cultured OL-SC or AC-SC groups, more neurites extended onto SCs compared with OLs and ACs. Conditioned media (CM) from OL or AC cultures did not reduce neurite length, implying that the inhibition of neurite growth by central glia is not due to soluble factors. Taken together, these results demonstrate that homogeneous populations of central glia inhibit SGN neurite growth.
Collapse
Affiliation(s)
- Eun-Ju Jeon
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa
- Department of Otolaryngology, Incheon St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Ningyong Xu
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa
| | - Lingjing Xu
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa
| | - Marlan R. Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa
| |
Collapse
|