351
|
Loers G, Aboul-Enein F, Bartsch U, Lassmann H, Schachner M. Comparison of myelin, axon, lipid, and immunopathology in the central nervous system of differentially myelin-compromised mutant mice: a morphological and biochemical study. Mol Cell Neurosci 2005; 27:175-89. [PMID: 15485773 DOI: 10.1016/j.mcn.2004.06.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2004] [Revised: 05/27/2004] [Accepted: 06/08/2004] [Indexed: 01/06/2023] Open
Abstract
The present study was carried out to compare different myelin-compromised mouse mutants with regard to myelin morphology in relation to axon-, lipid-, and immunopathology as a function of age. Mouse mutants deficient in the myelin-associated glycoprotein (MAG) and myelin basic protein (MBP) display subtle and severe myelin pathologies in the central nervous system (CNS), respectively. Animals doubly deficient in MAG and the neural cell adhesion molecule (NCAM) show defects similar to those present in MAG single mutants while mice deficient in MAG and the nonreceptor type tyrosine kinase Fyn are severely hypomyelinated, in addition to the MAG-specific myelin abnormalities. These mutant mice showed distinct myelin pathologies in different regions of the central nervous system and generally displayed a decrease in axonal integrity with age. Myelin pathology did not correlate locally with axon transection and with an involvement of the immune system as seen by numbers of CD3-positive lymphocytes and MAC-3-positive macrophages. Interestingly, the degree of these cellular abnormalities also did not correlate with abnormalities in levels of phospholipids, arachidonic acid, cholesterol, and apolipoprotein E (apoE). Moreover, these changes in lipid metabolism, including immune system-related arachidonic acid, preceded cellular pathology. The combined observations point to differences, but also similarities in the relation of myelin, axon, and immunopathology with genotype, and to a common aggravation of the phenotype with age.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universität Hamburg, D-20251 Hamburg, Germany
| | | | | | | | | |
Collapse
|
352
|
McPhail LT, Stirling DP, Tetzlaff W, Kwiecien JM, Ramer MS. The contribution of activated phagocytes and myelin degeneration to axonal retraction/dieback following spinal cord injury. Eur J Neurosci 2004; 20:1984-94. [PMID: 15450077 DOI: 10.1111/j.1460-9568.2004.03662.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Myelin-derived molecules inhibit axonal regeneration in the CNS. The Long-Evans Shaker rat is a naturally occurring dysmyelinated mutant, which although able to express the components of myelin lacks functional myelin in adulthood. Given that myelin breakdown exposes axons to molecules that are inhibitory to regeneration, we sought to determine whether injured dorsal column axons in a Shaker rat would exhibit a regenerative response absent in normally myelinated Long-Evans (control) rats. Although Shaker rat axons did not regenerate beyond the lesion, they remained at the caudal end of the crush site. Control rat axons, in contrast, retracted and died back from the edge of the crush. The absence of retraction/dieback in Shaker rats was associated with a reduced phagocytic reaction to dorsal column crush around the caudal edge of the lesion. Systemic injection of minocycline, a tetracycline derivative, in control rats reduced both the macrophage response and axonal retraction/dieback following dorsal column injury. In contrast, increasing macrophage activation by spinal injection of the yeast particulate zymosan had no effect on axonal retraction/dieback in Shaker rats. Schwann cell invasion was reduced in minocycline-treated control rats compared with untreated control rats, and was almost undetectable in Shaker rats, suggesting that like axonal retraction/dieback, spinal Schwann cell infiltration is dependent upon macrophage-mediated myelin degeneration. These results indicate that following spinal cord injury the phagocyte-mediated degeneration of myelin and subsequent exposure of inhibitory molecules to the injured axons contributes to their retraction/dieback.
Collapse
Affiliation(s)
- Lowell T McPhail
- ICORD (International Collaboration on Repair Discoveries), The University of British Columbia, 6270 University Boulevard, Vancouver, BC, Canada V6T 1Z4
| | | | | | | | | |
Collapse
|
353
|
Abstract
As neuronal development enters its final stages, axonal growth is restricted. Recent work indicates that several myelin-derived proteins, Nogo, MAG and OMgp, play a critical role in restricting axonal growth in the mature central nervous system (CNS). These proteins function by binding to an axonal NgR protein that limits axonal growth by activating Rho in neurons. Hypoxic conditions during the later stages of neuronal development have a prominent effect on oligodendrocytes and hence on the expression of these axon growth inhibitors. Reduced expression of these proteins caused by the developmental hypoxia, or direct blockade of the myelin inhibitor pathways in the adult CNS leads to axonal sprouting and the formation of new neuronal connections. The regulation of axonal growth, sprouting and connections in the postnatal brain by myelin proteins is an area of important investigation and potential therapeutic intervention.
Collapse
Affiliation(s)
- Fenghua Hu
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
354
|
Williams G, Williams EJ, Maison P, Pangalos MN, Walsh FS, Doherty P. Overcoming the inhibitors of myelin with a novel neurotrophin strategy. J Biol Chem 2004; 280:5862-9. [PMID: 15572360 DOI: 10.1074/jbc.m411121200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myelin inhibitors activate a p75(NTR)-dependent signaling cascade in neurons that not only inhibits axonal growth but also prevents neurotrophins (NT) from stimulating growth. Most intriguingly, in addition to Trk receptors, neurotrophins also bind to p75(NTR). We have designed a "mini-neurotrophin" called B(AG) to activate TrkB in the absence of p75(NTR) binding. We find that B(AG) is as effective as the natural TrkB ligands (brain-derived neurotrophic factor (BDNF) and NT-4) at promoting neurite outgrowth from cerebellar neurons. Furthermore, the neurite outgrowth responses stimulated by BDNF and B(AG) are inhibited by a common set of reagents, including the Trk receptor inhibitor K252a, as well as protein kinase A and phosphoinositide 3-kinase inhibitors. However, in contrast to BDNF, B(AG) promotes growth in the presence of a myelin inhibitor or when antibodies directly activate the p75(NTR) inhibitory pathway. On the basis of this observation, we postulated that the binding of BDNF to the p75(NTR) might compromise the ability of BDNF to stimulate neurite outgrowth in an inhibitory environment. To test this, we used NGF, and an NGF-derived peptide, to compete for the BDNF/p75(NTR) interaction; remarkably, in the presence of either agent, BDNF acquired the ability to promote neurite outgrowth in the presence of a myelin inhibitor. The data suggest that in an inhibitory environment, the BDNF/p75(NTR) interaction compromises regeneration. Agents that activate Trk receptors in the absence of p75(NTR) binding, or agents that inhibit neurotrophin/p75(NTR) binding, might therefore be better therapeutic candidates than neurotrophins.
Collapse
Affiliation(s)
- Gareth Williams
- The Wolfson Centre for Age-related Diseases, King's College London, London SE1 1UL, UK
| | | | | | | | | | | |
Collapse
|
355
|
Weiss J, Takizawa B, McGee A, Stewart WB, Zhang H, Ment L, Schwartz M, Strittmatter S. Neonatal hypoxia suppresses oligodendrocyte Nogo-A and increases axonal sprouting in a rodent model for human prematurity. Exp Neurol 2004; 189:141-9. [PMID: 15296844 DOI: 10.1016/j.expneurol.2004.05.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2004] [Revised: 04/08/2004] [Accepted: 05/14/2004] [Indexed: 11/16/2022]
Abstract
Premature human infants frequently suffer from periventricular leukomalacia (PVL) characterized by the loss of central myelinated tracts in the brain [Neuropathology, 22 (2002) 193]. Rodent chronic sublethal hypoxia (CSH) from P3 to 33 (postnatal day 3-33) provides a model for PVL characterized by cerebral ventriculomegaly and reductions in cerebral white matter volume [Brain Res. Dev. Brain Res. 111 (1998) 197; Proc. Natl. Acad. Sci. USA 100 (2003) 11718]. Here, we demonstrate that mice exposed to CSH from P3 to P33 followed by normoxia from P33 to P75 continue to exhibit a locomotor hyperactivity that resembles behavioral changes observed in some human children with very low birth weights. Because periventricular white matter is specifically lost in PVL, we examined the expression of oligodendrocyte proteins. Hypoxic rearing dramatically decreases the level of the axon outgrowth inhibitor Nogo-A in oligodendrocytes of CNS white matter at P12. The Nogo-A decrease exceeds the moderate decrease in another myelin protein, myelin associated glycoprotein (MAG). Although myelin protein expression returns to normal by maturity (P75), persistent abnormalities in axonal trajectories are detectable. Anterograde axonal tracing from motor cortex demonstrates ectopic corticofugal fibers in the corticospinal tract (CST), corpus callosum, and caudate nucleus of adult animals reared in CSH. Thus, hypoxia-induced reduction in myelin-derived axon outgrowth inhibitors appears to contribute axonal misconnection to the pathology of very low birth weight infants.
Collapse
Affiliation(s)
- Jared Weiss
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | |
Collapse
|
356
|
Li M, Shi J, Wei Z, Teng FYH, Tang BL, Song J. Structural characterization of the human Nogo-A functional domains. Solution structure of Nogo-40, a Nogo-66 receptor antagonist enhancing injured spinal cord regeneration. ACTA ACUST UNITED AC 2004; 271:3512-22. [PMID: 15317586 DOI: 10.1111/j.0014-2956.2004.04286.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The recent discovery of the Nogo family of myelin inhibitors and the Nogo-66 receptor opens up a very promising avenue for the development of therapeutic agents for treating spinal cord injury. Nogo-A, the largest member of the Nogo family, is a multidomain protein containing at least two regions responsible for inhibiting central nervous system (CNS) regeneration. So far, no structural information is available for Nogo-A or any of its structural domains. We have subcloned and expressed two Nogo-A fragments, namely the 182 residue Nogo-A(567-748) and the 66 residue Nogo-66 in Escherichia coli. CD and NMR characterization indicated that Nogo-A(567-748) was only partially structured while Nogo-66 was highly insoluble. Nogo-40, a truncated form of Nogo-66, has been previously shown to be a Nogo-66 receptor antagonist that is able to enhance CNS neuronal regeneration. Detailed NMR examinations revealed that a Nogo-40 peptide had intrinsic helix-forming propensity, even in an aqueous environment. The NMR structure of Nogo-40 was therefore determined in the presence of the helix-stabilizing solvent trifluoroethanol. The solution structure of Nogo-40 revealed two well-defined helices linked by an unstructured loop, representing the first structure of Nogo-66 receptor binding ligands. Our results provide the first structural insights into Nogo-A functional domains and may have implications in further designs of peptide mimetics that would enhance CNS neuronal regeneration.
Collapse
Affiliation(s)
- Minfen Li
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
357
|
Abstract
A hostile environment and decreased regenerative capacity may contribute to the failure of axon regeneration in the adult central nervous system. Recent studies leading to the identification of several myelin-associated inhibitors and their signaling molecules provide opportunitities to assess the contribution of these inhibitory molecules in restricting axon regeneration. These findings may ultimately allow for the development of strategies to alleviate the inhibitory effects of such molecules in an effort to encourage axon regeneration after spinal cord and brain injury.
Collapse
Affiliation(s)
- Zhigang He
- Division of Neuroscience, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
358
|
Yu W, Guo W, Feng L. Segregation of Nogo66 receptors into lipid rafts in rat brain and inhibition of Nogo66 signaling by cholesterol depletion. FEBS Lett 2004; 577:87-92. [PMID: 15527766 DOI: 10.1016/j.febslet.2004.09.068] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2004] [Revised: 09/16/2004] [Accepted: 09/16/2004] [Indexed: 11/20/2022]
Abstract
NogoA, a myelin-associated component, inhibits neurite outgrowth. Nogo66, a portion of NogoA, binds to Nogo66 receptor (NgR) and induces the inhibitory signaling. LINGO-1 and p75 neurotrophin receptor (p75), the low-affinity nerve growth factor receptor, are also required for NogoA signaling. However, signaling mechanisms downstream to Nogo receptor remain poorly understood. Here, we observed that NgR and p75 were colocalized in low-density membrane raft fractions derived from forebrains and cerebella as well as from cerebellar granule cells. NgR interacted with p75 in lipid rafts. In addition, disruption of lipid rafts by beta-methylcyclodextrin, a cholesterol-binding reagent, reduced the Nogo66 signaling. Our results suggest an important role of lipid rafts in facilitating the interaction between NgRs and provide insight into mechanisms underlying the inhibition of neurite outgrowth by NogoA.
Collapse
Affiliation(s)
- Weiying Yu
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, PR China
| | | | | |
Collapse
|
359
|
Kim JE, Liu BP, Park JH, Strittmatter SM. Nogo-66 Receptor Prevents Raphespinal and Rubrospinal Axon Regeneration and Limits Functional Recovery from Spinal Cord Injury. Neuron 2004; 44:439-51. [PMID: 15504325 DOI: 10.1016/j.neuron.2004.10.015] [Citation(s) in RCA: 270] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2004] [Revised: 08/30/2004] [Accepted: 09/21/2004] [Indexed: 10/26/2022]
Abstract
Axon regeneration after injury to the adult mammalian CNS is limited in part by three inhibitory proteins in CNS myelin: Nogo-A, MAG, and OMgp. All three of these proteins bind to a Nogo-66 receptor (NgR) to inhibit axonal outgrowth in vitro. To explore the necessity of NgR for responses to myelin inhibitors and for restriction of axonal growth in the adult CNS, we generated ngr(-/-) mice. Mice lacking NgR are viable but display hypoactivity and motor impairment. DRG neurons lacking NgR do not bind Nogo-66, and their growth cones are not collapsed by Nogo-66. Recovery of motor function after dorsal hemisection or complete transection of the spinal cord is improved in the ngr(-/-) mice. While corticospinal fibers do not regenerate in mice lacking NgR, regeneration of some raphespinal and rubrospinal fibers does occur. Thus, NgR is partially responsible for limiting the regeneration of certain fiber systems in the adult CNS.
Collapse
MESH Headings
- 5,7-Dihydroxytryptamine/pharmacology
- Animals
- Axons/physiology
- Behavior, Animal
- Blotting, Northern/methods
- Blotting, Southern/methods
- Brain/pathology
- Cell Count/methods
- Cells, Cultured
- Cloning, Molecular/methods
- Cornified Envelope Proline-Rich Proteins
- Desipramine/pharmacology
- Disease Models, Animal
- Evoked Potentials, Motor/physiology
- Female
- GPI-Linked Proteins
- Ganglia, Spinal/cytology
- Glial Fibrillary Acidic Protein/metabolism
- Glucose/analogs & derivatives
- Glucose/pharmacology
- Growth Cones/drug effects
- Growth Cones/physiology
- Immunohistochemistry/methods
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Motor Activity/physiology
- Myelin Proteins/genetics
- Myelin Proteins/metabolism
- Myelin Proteins/pharmacology
- Myelin Proteins/physiology
- Myelin Sheath/metabolism
- Myelin-Associated Glycoprotein
- Nerve Regeneration
- Neurons/cytology
- Neurons/drug effects
- Nogo Proteins
- Nogo Receptor 1
- Phospholipid Ethers/pharmacology
- Proteins/metabolism
- Pyramidal Tracts/physiopathology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/physiology
- Recovery of Function/physiology
- Serotonin/metabolism
- Serotonin Agents/pharmacology
- Spinal Cord/metabolism
- Spinal Cord/physiopathology
- Spinal Cord Injuries/genetics
- Spinal Cord Injuries/physiopathology
- Spinal Cord Injuries/prevention & control
- Time Factors
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
360
|
Schweigreiter R, Walmsley AR, Niederöst B, Zimmermann DR, Oertle T, Casademunt E, Frentzel S, Dechant G, Mir A, Bandtlow CE. Versican V2 and the central inhibitory domain of Nogo-A inhibit neurite growth via p75NTR/NgR-independent pathways that converge at RhoA. Mol Cell Neurosci 2004; 27:163-74. [PMID: 15485772 DOI: 10.1016/j.mcn.2004.06.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2004] [Accepted: 06/01/2004] [Indexed: 11/30/2022] Open
Abstract
Myelin is a major obstacle for regenerating nerve fibers of the adult mammalian central nervous system (CNS). Several proteins including Nogo-A, myelin-associated glycoprotein (MAG), oligodendrocyte myelin glycoprotein (OMgp) and the chondroitin-sulfate proteoglycan (CSPG) Versican V2 have been identified as inhibitory components present in CNS myelin. MAG, OMgp as well as the Nogo specific domain Nogo-66 exert their inhibitory activity by binding to a neuronal receptor complex containing the Nogo-66 receptor NgR and the neurotrophin receptor p75(NTR). While this suggests a converging role of the p75(NTR)/NgR receptor complex for myelin-derived neurite growth inhibitors, we show here that NgR/p75(NTR) is not required for mediating the inhibitory activity of the two myelin components NiG, unlike Nogo-66 a distinct domain of Nogo-A, and Versican V2. Primary neurons derived from a complete null mutant of p75(NTR) are still sensitive to NiG and Versican V2. In line with this result, neurite growth of p75(NTR) deficient neurons is still significantly blocked on total bovine CNS myelin. Furthermore, modulation of RhoA and Rac1 in p75(NTR)-/- neurons persists with NiG and Versican V2. Finally, we demonstrate that neither NiG nor Versican V2 interact with the p75(NTR)/NgR receptor complex and provide evidence that the binding sites of NiG and Nogo-66 are physically distinct from each other on neural tissue. These results indicate not only the existence of neuronal receptors for myelin inhibitors independent from the p75(NTR)/NgR receptor complex but also establish Rho GTPases as a common point of signal convergence of diverse myelin-induced regeneration inhibitory pathways.
Collapse
Affiliation(s)
- Rüdiger Schweigreiter
- Institute of Medical Chemistry and Biochemistry, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
361
|
Li W, Walus L, Rabacchi SA, Jirik A, Chang E, Schauer J, Zheng BH, Benedetti NJ, Liu BP, Choi E, Worley D, Silvian L, Mo W, Mullen C, Yang W, Strittmatter SM, Sah DWY, Pepinsky B, Lee DHS. A Neutralizing Anti-Nogo66 Receptor Monoclonal Antibody Reverses Inhibition of Neurite Outgrowth by Central Nervous System Myelin. J Biol Chem 2004; 279:43780-8. [PMID: 15297463 DOI: 10.1074/jbc.m401803200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Nogo66 receptor (NgR1) is a neuronal, leucine-rich repeat (LRR) protein that binds three central nervous system (CNS) myelin proteins, Nogo, myelin-associated glycoprotein, and oligodendrocyte myelin glycoprotein, and mediates their inhibitory effects on neurite growth. Although the LRR domains on NgR1 are necessary for binding to the myelin proteins, the exact epitope(s) involved in ligand binding is unclear. Here we report the generation and detailed characterization of an anti-NgR1 monoclonal antibody, 7E11. The 7E11 monoclonal antibody blocks Nogo, myelin-associated glycoprotein, and oligodendrocyte myelin glycoprotein binding to NgR1 with IC50 values of 120, 14, and 4.5 nm, respectively, and effectively promotes neurite outgrowth of P3 rat dorsal root ganglia neurons cultured on a CNS myelin substrate. Further, we have defined the molecular epitope of 7E11 to be DNAQLR located in the third LRR domain of rat NgR1. Our data demonstrate that anti-NgR1 antibodies recognizing this epitope, such as 7E11, can neutralize CNS myelin-dependent inhibition of neurite outgrowth. Thus, specific anti-NgR1 antibodies may represent a useful therapeutic approach for promoting CNS repair after injury.
Collapse
Affiliation(s)
- Weiwei Li
- Biogen Idec, Inc, Cambridge, Massachusetts 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
362
|
Abstract
Neuronal plasticity plays an important role in physiological and pathological processes within the gastrointestinal (GI) tract. Nogo A is a major contributor to the negative effect central nervous system (CNS) myelin has on neurite outgrowth after injury and may also play a role in maintaining synaptic connections in the healthy CNS. Nogo A is highly expressed during neuronal development but in the CNS declines postnatally concomitantly with a loss of regenerative potential while ganglia of the Peripheral Nervous System (PNS) retain Nogo A. The enteric nervous system shares a number of features in common with the CNS, thus the peripheral distribution of factors affecting plasticity is of interest. We have investigated the distribution of Nogo in the adult mammalian gastrointestinal tract. Nogo A mRNA and protein are detectable in the adult rat GI tract. Nogo A is expressed heterogeneously in enteric neurons throughout the GI tract though expression levels appear not to be correlated with neuronal sub-type. The pattern of expression is maintained in cultured myenteric plexus from the guinea-pig small intestine. As is seen in developing neurons of the CNS, enteric Nogo A is present in both neuronal cell bodies and axons. Our results point to a hitherto unsuspected role for Nogo A in enteric neuronal physiology.
Collapse
Affiliation(s)
- S L Osborne
- Neurology and Gastroenterology Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Science Park, Third Avenue, Harlow, Essex, UK
| | | | | | | |
Collapse
|
363
|
Rajagopalan S, Deitinghoff L, Davis D, Conrad S, Skutella T, Chedotal A, Mueller BK, Strittmatter SM. Neogenin mediates the action of repulsive guidance molecule. Nat Cell Biol 2004; 6:756-62. [PMID: 15258590 DOI: 10.1038/ncb1156] [Citation(s) in RCA: 219] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Accepted: 06/21/2004] [Indexed: 11/08/2022]
Abstract
Repulsive guidance molecule (RGM) is a recently identified protein implicated in both axonal guidance and neural tube closure. The avoidance of chick RGM in the posterior optic tectum by growing temporal, but not nasal, retinal ganglion cell axons is thought to contribute to visual map formation. In contrast to ephrins, semaphorins, netrins and slits, no receptor mechanism for RGM action has been defined. Here, an expression cloning strategy identified neogenin as a binding site for RGM, with a sub-nanomolar affinity. Consistent with selective axonal responsiveness to RGM, neogenin is expressed in a gradient across the chick retina. Neogenin is known to be one of several netrin-binding proteins but only neogenin interacts with RGM. The avoidance of RGM by temporal retinal axons is blocked by the anti-neogenin antibody and the soluble neogenin ectodomain. Dorsal root ganglion axons are unresponsive to RGM but are converted to a responsive state by neogenin expression. Thus, neogenin functions as an RGM receptor.
Collapse
Affiliation(s)
- Srikanth Rajagopalan
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | |
Collapse
|
364
|
Nikulina E, Tidwell JL, Dai HN, Bregman BS, Filbin MT. The phosphodiesterase inhibitor rolipram delivered after a spinal cord lesion promotes axonal regeneration and functional recovery. Proc Natl Acad Sci U S A 2004; 101:8786-90. [PMID: 15173585 PMCID: PMC423273 DOI: 10.1073/pnas.0402595101] [Citation(s) in RCA: 261] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2004] [Indexed: 01/24/2023] Open
Abstract
Although there is no spontaneous regeneration of mammalian spinal axons after injury, they can be enticed to grow if cAMP is elevated in the neuronal cell bodies before the spinal axons are cut. Prophylactic injection of cAMP, however, is useless as therapy for spinal injuries. We now show that the phosphodiesterase 4 (PDE4) inhibitor rolipram (which readily crosses the blood-brain barrier) overcomes inhibitors of regeneration in myelin in culture and promotes regeneration in vivo. Two weeks after a hemisection lesion at C3/4, with embryonic spinal tissue implanted immediately at the lesion site, a 10-day delivery of rolipram results in considerable axon regrowth into the transplant and a significant improvement in motor function. Surprisingly, in rolipram-treated animals, there was also an attenuation of reactive gliosis. Hence, because rolipram promotes axon regeneration, attenuates the formation of the glial scar, and significantly enhances functional recovery, and because it is effective when delivered s.c., as well as post-injury, it is a strong candidate as a useful therapy subsequent to spinal cord injury.
Collapse
Affiliation(s)
- Elena Nikulina
- Biology Department, Hunter College, City University of New York, New York, NY 10024, USA
| | | | | | | | | |
Collapse
|
365
|
Sandvig A, Berry M, Barrett LB, Butt A, Logan A. Myelin-, reactive glia-, and scar-derived CNS axon growth inhibitors: expression, receptor signaling, and correlation with axon regeneration. Glia 2004; 46:225-51. [PMID: 15048847 DOI: 10.1002/glia.10315] [Citation(s) in RCA: 286] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Axon regeneration is arrested in the injured central nervous system (CNS) by axon growth-inhibitory ligands expressed in oligodendrocytes/myelin, NG2-glia, and reactive astrocytes in the lesion and degenerating tracts, and by fibroblasts in scar tissue. Growth cone receptors (Rc) bind inhibitory ligands, activating a Rho-family GTPase intracellular signaling pathway that disrupts the actin cytoskeleton inducing growth cone collapse/repulsion. The known inhibitory ligands include the chondroitin sulfate proteoglycans (CSPG) Neurocan, Brevican, Phosphacan, Tenascin, and NG2, as either membrane-bound or secreted molecules; Ephrins expressed on astrocyte/fibroblast membranes; the myelin/oligodendrocyte-derived growth inhibitors Nogo, MAG, and OMgp; and membrane-bound semaphorins (Sema) produced by meningeal fibroblasts invading the scar. No definitive CSPG Rc have been identified, although intracellular signaling through the Rho family of G-proteins is probably common to all the inhibitory ligands. Ephrins bind to signalling Ephs. The ligand-binding Rc for all the myelin inhibitors is NgR and requires p75(NTR) for transmembrane signaling. The neuropilin (NP)/plexin (Plex) Rc complex binds Sema. Strategies for promoting axon growth after CNS injury are thwarted by the plethora of inhibitory ligands and the ligand promiscuity of some of their Rc. There is also paradoxical reciprocal expression of many of the inhibitory ligands/Rc in normal and damaged neurons, and NgR expression is restricted to a limited number of neuronal populations. All these factors, together with an incomplete understanding of the normal functions of many of these molecules in the intact CNS, presently confound interpretive acumen in regenerative studies.
Collapse
Affiliation(s)
- Axel Sandvig
- Laboratory of Regenerative Neurobiology, Institute for Experimental Medical Research, Ullevål University Hospital, Oslo, Norway.
| | | | | | | | | |
Collapse
|
366
|
Profyris C, Cheema SS, Zang D, Azari MF, Boyle K, Petratos S. Degenerative and regenerative mechanisms governing spinal cord injury. Neurobiol Dis 2004; 15:415-36. [PMID: 15056450 DOI: 10.1016/j.nbd.2003.11.015] [Citation(s) in RCA: 347] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2003] [Revised: 11/03/2003] [Accepted: 11/14/2003] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is a major cause of disability, and at present, there is no universally accepted treatment. The functional decline following SCI is contributed to both direct mechanical injury and secondary pathophysiological mechanisms that are induced by the initial trauma. These mechanisms initially involve widespread haemorrhage at the site of injury and necrosis of central nervous system (CNS) cellular components. At later stages of injury, the cord is observed to display reactive gliosis. The actions of astrocytes as well as numerous other cells in this response create an environment that is highly nonpermissive to axonal regrowth. Also manifesting important effects is the immune system. The early recruitment of neutrophils and at later stages, macrophages to the site of insult cause exacerbation of injury. However, at more chronic stages, macrophages and recruited T helper cells may potentially be helpful by providing trophic support for neuronal and non-neuronal components of the injured CNS. Within this sea of injurious mechanisms, the oligodendrocytes appear to be highly vulnerable. At chronic stages of SCI, a large number of oligodendrocytes undergo apoptosis at sites that are distant to the vicinity of primary injury. This leads to denudement of axons and deterioration of their conductive abilities, which adds significantly to functional decline. By indulging into the molecular mechanisms that cause oligodendrocyte apoptosis and identifying potential targets for therapeutic intervention, the prevention of this apoptotic wave will be of tremendous value to individuals living with SCI.
Collapse
Affiliation(s)
- Christos Profyris
- Motor Neuron Disease and Paralysis Laboratory, Neural Injury and Repair Group, The Howard Florey Institute of Experimental Physiology and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | |
Collapse
|
367
|
Lie DC, Song H, Colamarino SA, Ming GL, Gage FH. Neurogenesis in the adult brain: new strategies for central nervous system diseases. Annu Rev Pharmacol Toxicol 2004; 44:399-421. [PMID: 14744252 DOI: 10.1146/annurev.pharmtox.44.101802.121631] [Citation(s) in RCA: 441] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
New cells are continuously generated from immature proliferating cells throughout adulthood in many organs, thereby contributing to the integrity of the tissue under physiological conditions and to repair following injury. In contrast, repair mechanisms in the adult central nervous system (CNS) have long been thought to be very limited. However, recent findings have clearly demonstrated that in restricted areas of the mammalian brain, new functional neurons are constantly generated from neural stem cells throughout life. Moreover, stem cells with the potential to give rise to new neurons reside in many different regions of the adult CNS. These findings raise the possibility that endogenous neural stem cells can be mobilized to replace dying neurons in neurodegenerative diseases. Indeed, recent reports have provided evidence that, in some injury models, limited neuronal replacement occurs in the CNS. Here, we summarize our current understanding of the mechanisms controlling adult neurogenesis and discuss their implications for the development of new strategies for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- D Chichung Lie
- Laboratory of Genetics, The Salk Institute, La Jolla, California 92037, USA.
| | | | | | | | | |
Collapse
|
368
|
Laurén J, Airaksinen MS, Saarma M, Timmusk T. Two novel mammalian Nogo receptor homologs differentially expressed in the central and peripheral nervous systems. Mol Cell Neurosci 2004; 24:581-94. [PMID: 14664809 DOI: 10.1016/s1044-7431(03)00199-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The regenerative capacity of the adult mammalian central nervous system is restricted by the myelinating oligodendrocytes that form a nonpermissive environment for axonal growth. Currently only the Nogo receptor (NgR), in complex with p75(NTR) neurotrophin receptor is known to be involved in this inhibitory signalling in neurons. NgR is a common receptor for the three inhibitory myelin proteins Nogo-A, OMgp, and MAG. Here we describe two novel Nogo receptor gene homologs named NGRL2 and NGRL3 from human and mouse that, like NGR, encode putative leucine-rich repeat containing GPI-anchored proteins. We show by in situ hybridisation and by RT-PCR that NGRL mRNAs are predominantly expressed in the neurons of the embryonic and adult central and peripheral nervous systems, and that they together with NGR possess distinct and partially nonoverlapping expression patterns. We also show that all four members of the reticulon family, including Nogo-A, are widely expressed in the nervous system, and therefore are possible ligands for the NgRLs.
Collapse
MESH Headings
- Amino Acid Sequence/genetics
- Animals
- Base Sequence/genetics
- Central Nervous System/cytology
- Central Nervous System/growth & development
- Central Nervous System/metabolism
- DNA, Complementary/analysis
- DNA, Complementary/genetics
- Fetus
- GPI-Linked Proteins
- Gene Expression Regulation, Developmental/genetics
- Ligands
- Membrane Proteins/genetics
- Mice
- Molecular Sequence Data
- Muscle Proteins/genetics
- Myelin Proteins/metabolism
- Nerve Regeneration/physiology
- Nerve Tissue Proteins/genetics
- Nogo Proteins
- Nogo Receptor 1
- Oligodendroglia/metabolism
- Peripheral Nervous System/cytology
- Peripheral Nervous System/growth & development
- Peripheral Nervous System/metabolism
- RNA, Messenger/metabolism
- Receptors, Cell Surface
- Receptors, Peptide/biosynthesis
- Receptors, Peptide/genetics
- Receptors, Peptide/isolation & purification
- Sequence Homology, Amino Acid
- Sequence Homology, Nucleic Acid
Collapse
Affiliation(s)
- Juha Laurén
- Program in Molecular Neurobiology, Institute of Biotechnology, University of Helsinki, P.O. Box 56 (Viikinkaari 9), FIN-00014, University of Helsinki, Helsinki, Finland.
| | | | | | | |
Collapse
|
369
|
Sapieha PS, Peltier M, Rendahl KG, Manning WC, Di Polo A. Fibroblast growth factor-2 gene delivery stimulates axon growth by adult retinal ganglion cells after acute optic nerve injury. Mol Cell Neurosci 2004; 24:656-72. [PMID: 14664816 DOI: 10.1016/s1044-7431(03)00228-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Basic fibroblast growth factor (or FGF-2) has been shown to be a potent stimulator of retinal ganglion cell (RGC) axonal growth during development. Here we investigated if FGF-2 upregulation in adult RGCs promoted axon regrowth in vivo after acute optic nerve injury. Recombinant adeno-associated virus (AAV) was used to deliver the FGF-2 gene to adult RGCs providing a sustained source of this neurotrophic factor. FGF-2 gene transfer led to a 10-fold increase in the number of axons that extended past 0.5 mm from the lesion site compared to control nerves. Detection of AAV-mediated FGF-2 protein in injured RGC axons correlated with growth into the distal optic nerve. The response to FGF-2 upregulation was supported by our finding that FGF receptor-1 (FGFR-1) and heparan sulfate (HS), known to be essential for FGF-2 signaling, were expressed by adult rat RGCs. FGF-2 transgene expression led to only transient protection of injured RGCs. Thus the effect of this neurotrophic factor on axon extension could not be solely attributed to an increase in neuronal survival. Our data indicate that selective upregulation of FGF-2 in adult RGCs stimulates axon regrowth within the optic nerve, an environment that is highly inhibitory for regeneration. These results support the hypothesis that key factors involved in axon outgrowth during neural development may promote regeneration of adult injured neurons.
Collapse
Affiliation(s)
- Przemyslaw S Sapieha
- Department of Pathology and Cell Biology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | | | | | | | | |
Collapse
|
370
|
Fischer D, He Z, Benowitz LI. Counteracting the Nogo receptor enhances optic nerve regeneration if retinal ganglion cells are in an active growth state. J Neurosci 2004; 24:1646-51. [PMID: 14973241 PMCID: PMC6730473 DOI: 10.1523/jneurosci.5119-03.2004] [Citation(s) in RCA: 209] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mature retinal ganglion cells (RGCs), like other CNS neurons, cannot regrow injured axons into a myelin-rich environment. If stimulated by macrophage-derived factors, however, RGCs can regenerate their axons for considerable distances through the distal optic nerve. Using this "sensitized background," we investigated the effects of either increasing the expression or suppressing the activity of the Nogo receptor (NgR). NgR mediates the growth-inhibiting effects of three myelin proteins, Nogo, OMgp (oligodendrocyte-myelin glycoprotein), and MAG (myelin-associated glycoprotein). Transfecting growth-sensitized RGCs with adeno-associated viruses expressing a dominant-negative form of NgR (NgR(DN)) increased axon regeneration several-fold; however, when the growth program of RGCs was not activated, NgR(DN) expression had no beneficial effects. Overexpression of wild-type NgR blocked almost all regeneration from growth-sensitized RGCs and caused axons proximal to the lesion site to retract. We conclude that gene therapy is an effective approach to enhancing axon regeneration in the CNS and that inactivation of NgR functioning greatly enhances axon regeneration provided the intrinsic growth program of neurons is activated.
Collapse
Affiliation(s)
- Dietmar Fischer
- Department of Neurosurgery, Children's Hospital Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
371
|
Kretz A, Kügler S, Happold C, Bähr M, Isenmann S. Excess Bcl-XL increases the intrinsic growth potential of adult CNS neurons in vitro. Mol Cell Neurosci 2004; 26:63-74. [PMID: 15121179 DOI: 10.1016/j.mcn.2004.01.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2003] [Revised: 01/15/2004] [Accepted: 01/16/2004] [Indexed: 02/01/2023] Open
Abstract
The regenerative potential of adult mammalian CNS neurons is limited. Recent data suggest that inactivation of major growth inhibitors may not suffice to induce robust regeneration from mature neurons unless the intrinsic growth state is modulated. To investigate a possible role of Bcl-XL for axon regeneration in the adult mammalian CNS, Bcl-XL was adenovirally overexpressed in severed rat RGCs. Bcl-XL overexpression in mature axotomized RGCs in vivo increased both numbers [3.10-fold (+/-0.20)] and cumulative length [6.72-fold (+/-0.47)] of neurites regenerated from retinal explants, and this effect was further pronounced in the central retina where specific and dense axoplasmatic transduction occurs. Similarly, delayed Bcl-XL gene transfer to explanted retinae 12-13 days after lesion increased the numbers and length of emanating neurites by a factor of 5.22 (+/-0.41) and 8.29 (+/-0.69), respectively. In vivo, intraretinal sprouting of unmyelinated RGC axons into the nerve fiber layer was increased. However, fiber ingrowth into the optic nerve remained sparse, likely due to myelin inhibitors and scar components. Therefore, Bcl-XL overexpression may enhance, but may not be sufficient to, restitute functional regeneration in the adult CNS. As assessed by cell quantification analysis, Bcl-XL overexpression rescued a higher proportion of RGCs in vivo than in vitro. Therefore, Bcl-XL is capable to induce both neuronal survival and axon regeneration, but these two processes appear to be differentially modified by distinct pathways in vivo.
Collapse
Affiliation(s)
- Alexandra Kretz
- Neuroregeneration Laboratory, Department of Neurology, University of Jena Medical School, Jena, Germany
| | | | | | | | | |
Collapse
|
372
|
Vourc'h P, Andres C. Oligodendrocyte myelin glycoprotein (OMgp): evolution, structure and function. ACTA ACUST UNITED AC 2004; 45:115-24. [PMID: 15145622 DOI: 10.1016/j.brainresrev.2004.01.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2004] [Indexed: 12/16/2022]
Abstract
The oligodendrocyte myelin glycoprotein (OMgp) is a glycosylphosphatidylinositol-anchored protein expressed by neurons and oligodendrocytes in the central nervous system (CNS). Although the precise function of OMgp is yet to be determined in vivo, recent in vitro studies suggested roles for this protein in both the developing and adult central nervous system. In vitro experiments demonstrated the participation of OMgp in growth cone collapse and inhibition of neurite outgrowth through its interaction with NgR, the receptor for Nogo. This function requires its leucine-rich repeat domain, a highly conserved region in OMgp during mammal evolution. OMgp leucine-rich repeat domain is also implicated in the inhibition of cell proliferation. Based on its developmental expression, localization and structure, OMgp may also be involved in the formation and maintenance of myelin sheaths. Cell proliferation, neuronal sprouting and myelination are crucial processes involved in brain development and regeneration after injury. Here, we review the information available on the structure and evolution of OMgp, summarize its tissue expression and discuss its putative role(s) during the development and in adult CNS.
Collapse
Affiliation(s)
- Patrick Vourc'h
- Génétique et physiopathologie de l'autisme et des déficiences mentales, INSERM U619, CHRU Tours and Faculté de Médecine, 2 bis Bd Tonnellé, 37032 Tours Cedex, France
| | | |
Collapse
|
373
|
Cui Q, Cho KS, So KF, Yip HK. Synergistic effect of Nogo-neutralizing antibody IN-1 and ciliary neurotrophic factor on axonal regeneration in adult rodent visual systems. J Neurotrauma 2004; 21:617-25. [PMID: 15165369 DOI: 10.1089/089771504774129946] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The presence of Nogo axon regeneration inhibitory molecules in the central nervous system (CNS) and the counteracting effect of IN-1 antibodies have been widely reported. In this study, we examined the effect of IN-1-producing hybridoma cells on axon regeneration in adult rodent retinal ganglion cells (RGCs) after various types of optic nerve (ON) injury, evaluating therein whether ciliary neurotrophic factor (CNTF) potentiated the effect of IN-1. We found that application of IN-1 alone failed to enhance regeneration of intracranially or intraorbitally transected RGC axons in a peripheral nerve (PN) graft. IN-1 hybridoma cells also failed to significantly promote intraorbitally crushed ON axons to reenter the distal part of the ON. However, a combined application of IN-1 and CNTF had a synergistic effect in both intracranial PN and intraorbital ON crush paradigms. This study suggests that the action of IN-1 antibodies in promoting axon regeneration in the CNS could be more effective when coupled with other appropriate factors.
Collapse
Affiliation(s)
- Qi Cui
- Laboratory for Neural Repair, Shantou University Medical College, Shantou, China
| | | | | | | |
Collapse
|
374
|
Taylor JSH, Bampton ETW. Factors secreted by Schwann cells stimulate the regeneration of neonatal retinal ganglion cells. J Anat 2004; 204:25-31. [PMID: 14690475 PMCID: PMC1571234 DOI: 10.1111/j.1469-7580.2004.00262.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The adult mammalian central nervous system (CNS) does not repair after injury. However, we and others have shown in earlier work that the neonatal CNS is capable of repair and importantly of allowing regenerating axons to re-navigate through the same pathways as they did during development. This phase of neonatal repair is restricted by the fragility of neurons after injury and a lack of trophic factors that enable their survival. Our aim is to define better the factors that sustain neurons after injury and allow regeneration to occur. We describe some of our work using Schwann cells to promote the regeneration of neurons from young postnatal rodents. We have established rapid methods for purifying Schwann cells without the use of either anti-mitotic agents to suppress contaminating fibroblasts or mitotic stimulation to generate large numbers of Schwann cells. The rapidly purified Schwann cells have been used to generate conditioned medium that we have shown stimulates axon regeneration in cultured retinal ganglion cell neurons. We also show that the positive effects of Schwann cells are still present after pharmacological blockade of the neurotrophin receptors, suggesting that novel factors mediate these effects.
Collapse
|
375
|
Abstract
Injury to the adult mammalian central nervous system (CNS) often results in permanent loss of sensory and motor function. This is due to the failure of injured axons to regenerate. The inhibitory nature of the CNS can be attributed to several factors, including formation of the glial scar, the presence of several molecules, associated with myelin, which inhibit axonal regrowth, and the intrinsic growth state of these neurons. Encouraging regeneration in the adult mammalian CNS therefore will require targeting one or all of these factors following injury. Here we illustrate recent work from our laboratory that identifies some of the signalling components involved in modulation of the intrinsic growth state of adult neurons. When activated, these signalling pathways can induce axonal regeneration in the presence of the myelin-associated inhibitors both in vitro and in vivo.
Collapse
Affiliation(s)
- Tim Spencer
- Department of Biological Sciences, Hunter College, The City University of New York, NY 10021, USA
| | | |
Collapse
|
376
|
Mi S, Lee X, Shao Z, Thill G, Ji B, Relton J, Levesque M, Allaire N, Perrin S, Sands B, Crowell T, Cate RL, McCoy JM, Pepinsky RB. LINGO-1 is a component of the Nogo-66 receptor/p75 signaling complex. Nat Neurosci 2004; 7:221-8. [PMID: 14966521 DOI: 10.1038/nn1188] [Citation(s) in RCA: 637] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2003] [Accepted: 12/24/2003] [Indexed: 02/02/2023]
Abstract
Axon regeneration in the adult CNS is prevented by inhibitors in myelin. These inhibitors seem to modulate RhoA activity by binding to a receptor complex comprising a ligand-binding subunit (the Nogo-66 receptor NgR1) and a signal transducing subunit (the neurotrophin receptor p75). However, in reconstituted non-neuronal systems, NgR1 and p75 together are unable to activate RhoA, suggesting that additional components of the receptor may exist. Here we describe LINGO-1, a nervous system-specific transmembrane protein that binds NgR1 and p75 and that is an additional functional component of the NgR1/p75 signaling complex. In non-neuronal cells, coexpression of human NgR1, p75 and LINGO-1 conferred responsiveness to oligodendrocyte myelin glycoprotein, as measured by RhoA activation. A dominant-negative human LINGO-1 construct attenuated myelin inhibition in transfected primary neuronal cultures. This effect on neurons was mimicked using an exogenously added human LINGO-1-Fc fusion protein. Together these observations suggest that LINGO-1 has an important role in CNS biology.
Collapse
MESH Headings
- Amino Acid Sequence/genetics
- Animals
- Animals, Newborn
- Astrocytes/metabolism
- Axons/metabolism
- Base Sequence/genetics
- Cells, Cultured
- DNA, Complementary/analysis
- DNA, Complementary/genetics
- Fetus
- GPI-Linked Proteins
- Humans
- Macromolecular Substances
- Membrane Proteins/genetics
- Membrane Proteins/isolation & purification
- Membrane Proteins/metabolism
- Molecular Sequence Data
- Mutation/genetics
- Myelin Proteins/metabolism
- Myelin Sheath/metabolism
- Myelin-Associated Glycoprotein/genetics
- Myelin-Associated Glycoprotein/isolation & purification
- Myelin-Associated Glycoprotein/metabolism
- Myelin-Oligodendrocyte Glycoprotein
- Nerve Regeneration/physiology
- Nerve Tissue Proteins
- Nogo Receptor 1
- Protein Structure, Tertiary/genetics
- Rats
- Receptor, Nerve Growth Factor
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/isolation & purification
- Receptors, Cell Surface/metabolism
- Receptors, Nerve Growth Factor/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/genetics
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Sha Mi
- Department of Discovery Biology, Biogen Idec, Inc., 14 Cambridge Center, Cambridge, Massachusetts 02142, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
377
|
Sivasankaran R, Pei J, Wang KC, Zhang YP, Shields CB, Xu XM, He Z. PKC mediates inhibitory effects of myelin and chondroitin sulfate proteoglycans on axonal regeneration. Nat Neurosci 2004; 7:261-8. [PMID: 14770187 DOI: 10.1038/nn1193] [Citation(s) in RCA: 242] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2003] [Accepted: 01/12/2004] [Indexed: 01/18/2023]
Abstract
Successful axon regeneration in the mammalian central nervous system (CNS) is at least partially compromised due to the inhibitors associated with myelin and glial scar. However, the intracellular signaling mechanisms underlying these inhibitory activities are largely unknown. Here we provide biochemical and functional evidence that conventional isoforms of protein kinase C (PKC) are key components in the signaling pathways that mediate the inhibitory activities of myelin components and chondroitin sulfate proteoglycans (CSPGs), the major class of inhibitors in the glial scar. Both the myelin inhibitors and CSPGs induce PKC activation. Blocking PKC activity pharmacologically and genetically attenuates the ability of CNS myelin and CSPGs to activate Rho and inhibit neurite outgrowth. Intrathecal infusion of a PKC inhibitor, Gö6976, into the site of dorsal hemisection promotes regeneration of dorsal column axons across and beyond the lesion site in adult rats. Thus, perturbing PKC activity could represent a therapeutic approach to stimulating axon regeneration after brain and spinal cord injuries.
Collapse
Affiliation(s)
- Rajeev Sivasankaran
- Division of Neuroscience, 320 Longwood Avenue, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
378
|
Marta CB, Taylor CM, Cheng S, Quarles RH, Bansal R, Pfeiffer SE. Myelin associated glycoprotein cross-linking triggers its partitioning into lipid rafts, specific signaling events and cytoskeletal rearrangements in oligodendrocytes. NEURON GLIA BIOLOGY 2004; 1:35-46. [PMID: 16998591 PMCID: PMC1570761 DOI: 10.1017/s1740925x04000067] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Myelin-associated glycoprotein (MAG) has been implicated in inhibition of nerve regeneration in the CNS. This results from interactions between MAG and the Nogo receptor and gangliosides on the apposing axon, which generates intracellular inhibitory signals in the neuron. However, because myelin-axon signaling is bidirectional, we undertook an analysis of potential MAG-activated signaling in oligodendrocytes (OLs). In this study, we show that antibody cross-linking of MAG on the surface of OLs (to mimic axonal binding) leads to the redistribution of MAG into detergent (TX-100)-insoluble complexes, hyperphosphorylation of Fyn, dephosphorylation of serine and threonine residues in specific proteins, including lactate dehydrogenase and the beta subunit of the trimeric G-protein-complex, and cleavage of alpha-fodrin followed by a transient depolymerization of actin. We propose that these changes are part of a signaling cascade in OLs associated with MAG function as a mediator of axon-glial communication which might have implications for the mutual regulation of the formation and stability of axons and myelin.
Collapse
Affiliation(s)
- C B Marta
- Department of Neuroscience, University of Connecticut Medical School, 263 Farmington Avenue, Farmington, CT 06030-3401, USA.
| | | | | | | | | | | |
Collapse
|
379
|
Neurotrophins elevate cAMP to reach a threshold required to overcome inhibition by MAG through extracellular signal-regulated kinase-dependent inhibition of phosphodiesterase. J Neurosci 2004. [PMID: 14684879 DOI: 10.1523/jneurosci.23-37-11770.2003] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inhibitors of regeneration in myelin, such as myelin-associated glycoprotein (MAG), play an important role in preventing regeneration after CNS injury. Elevation of cAMP, either with dibutyryl-cAMP (db-cAMP) or by priming with a variety of neurotrophins, overcomes inhibition by MAG and myelin. However, activation of cAMP is not generally regarded as a signaling pathway for neurotrophins. Here we show that the NGF-like neurotrophins overcome inhibition by MAG by activating tyrosine kinase receptors. We also show that activation of extracellular signal-regulated kinase (Erk) by BDNF is required to overcome inhibition by MAG, and that activated Erk transiently inhibits phosphodiesterase 4 (PDE4), the enzyme that hydrolyzes cAMP. Inhibition of PDE4 then allows cAMP to increase and so initiates the pathway to overcome inhibition. Furthermore, we also show that basal levels of Erk activation and basal cAMP levels contribute to the effects of db-cAMP by pushing the combined levels of cAMP above a threshold required to overcome inhibition. Together, these results not only show how NGF-like neurotrophins can elevate cAMP and overcome inhibition but also point to a novel mechanism of cross talk in neurons from the Erk to the cAMP signaling pathways.
Collapse
|
380
|
Lee DHS, Strittmatter SM, Sah DWY. Targeting the Nogo receptor to treat central nervous system injuries. Nat Rev Drug Discov 2004; 2:872-8. [PMID: 14668808 DOI: 10.1038/nrd1228] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Daniel H S Lee
- Biogen Inc., 14 Cambridge Center, Cambridge, Massachusetts 02142, USA.
| | | | | |
Collapse
|
381
|
Menon K, Rasband MN, Taylor CM, Brophy P, Bansal R, Pfeiffer SE. The myelin-axolemmal complex: biochemical dissection and the role of galactosphingolipids. J Neurochem 2004; 87:995-1009. [PMID: 14622129 DOI: 10.1046/j.1471-4159.2003.02075.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Myelin-axolemmal interactions regulate many cellular and molecular events, including gene expression, oligodendrocyte survival and ion channel clustering. Here we report the biochemical fractionation and enrichment of distinct subcellular domains from myelinated nerve fibers. Using antibodies against proteins found in compact myelin, non-compact myelin and axolemma, we show that a rigorous procedure designed to purify myelin also results in the isolation of the myelin-axolemmal complex, a high-affinity protein complex consisting of axonal and oligodendroglial components. Further, the isolation of distinct subcellular domains from galactolipid-deficient mice with disrupted axoglial junctions is altered in a manner consistent with the delocalization of axolemmal proteins observed in these animals. These results suggest a paradigm for identification of proteins involved in neuroglial signaling.
Collapse
Affiliation(s)
- Krishna Menon
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut 06030-3401, USA
| | | | | | | | | | | |
Collapse
|
382
|
Klinger M, Taylor JS, Oertle T, Schwab ME, Stuermer CAO, Diekmann H. Identification of Nogo-66 receptor (NgR) and homologous genes in fish. Mol Biol Evol 2004; 21:76-85. [PMID: 12949137 DOI: 10.1093/molbev/msg241] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Nogo-66 receptor NgR has been implicated in the mediation of inhibitory effects of central nervous system (CNS) myelin on axon growth in the adult mammalian CNS. NgR binds to several myelin-associated ligands (Nogo-66, myelin associated glycoprotein, and oligodendrocyte-myelin glycoprotein), which, among other inhibitory proteins, impair axonal regeneration in the CNS of adult mammals. In contrast to mammals, severed axons readily regenerate in the fish CNS. Nevertheless, fish axons are repelled by mammalian oligodendrocytes in vitro. Therefore, the identification of fish NgR homologs is a crucial step towards understanding NgR functions in vertebrate systems competent of CNS regeneration. Here, we report the discovery of four zebrafish (Danio rerio) and five fugu (Takifugu rubripes) NgR homologs. Synteny between fish and human, comparable intron-exon structures, and phylogenetic analyses provide convincing evidence that the true fish orthologs were identified. The topology of the phylogenetic trees shows that the extra fish genes were produced by duplication events that occurred in ray-finned fishes before the divergence of the zebrafish and pufferfish lineages. Expression of zebrafish NgR homologs was detected relatively early in development and prominently in the adult brain, suggesting functions in axon growth, guidance, or plasticity.
Collapse
Affiliation(s)
- Michael Klinger
- Department of Biology, University of Konstanz, Konstanz, Germany
| | | | | | | | | | | |
Collapse
|
383
|
Reep RL, Cheatwood JL, Corwin JV. Role of the Dorsocentral Striatum in Contralateral Neglect and Recovery from Neglect in Rats. BIOCOMPUTING 2004. [DOI: 10.1007/978-1-4613-0225-4_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
384
|
Hasegawa Y, Yamagishi S, Fujitani M, Yamashita T. p75 neurotrophin receptor signaling in the nervous system. BIOTECHNOLOGY ANNUAL REVIEW 2004; 10:123-49. [PMID: 15504705 DOI: 10.1016/s1387-2656(04)10005-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The neurotrophin receptor p75(NTR) has long been known as a receptor for neurotrophins that promote survival and differentiation. Consistent with the role of neurotrophins, p75(NTR) is expressed during the developmental stages of the nervous system. However, p75(NTR) is re-expressed in various pathological conditions in the adult. We now know that p75(NTR) has the ability to elicit bi-directional signals, that result in the inhibition as well as the promotion of the neurite outgrowth. p75(NTR) is a key receptor for myelin-derived inhibitory cues that contribute to the lack of regeneration of the central nervous system.
Collapse
Affiliation(s)
- Yuiko Hasegawa
- Department of Neurobiology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | | | | | | |
Collapse
|
385
|
Abstract
Axon growth inhibitors associated with myelin and the glial scar contribute to the failure of axon regeneration in the injured adult mammalian central nervous system (CNS). A number of these inhibitors, their receptors, and signaling pathways have been identified. These inhibitors can now be neutralized by a variety of approaches that point to the possibility of developing new therapeutic strategies to stimulate regeneration after spinal cord injury.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, McGill University Health Centre, Montreal General Hospital Research Institute, 1650 Cedar Avenue, Montreal, Quebec, Canada, H3G 1A4.
| | | |
Collapse
|
386
|
Huber AB, Kolodkin AL, Ginty DD, Cloutier JF. Signaling at the growth cone: ligand-receptor complexes and the control of axon growth and guidance. Annu Rev Neurosci 2003; 26:509-63. [PMID: 12677003 DOI: 10.1146/annurev.neuro.26.010302.081139] [Citation(s) in RCA: 570] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The guidance of axons during the establishment of the nervous system is mediated by a variety of extracellular cues that govern cytoskeletal dynamics in axonal growth cones. A large number of these guidance cues and their cell-surface receptors have now been identified, and the intracellular signaling pathways by which these cues induce cytoskeletal rearrangements are becoming defined. This review summarizes our current understanding of the major families of axon guidance cues and their receptors, with a particular emphasis on receptor signaling mechanisms. We also discuss recent advances in understanding receptor cross talk and how the activities of guidance cues and their receptors are modulated during neural development.
Collapse
Affiliation(s)
- Andrea B Huber
- Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
387
|
Abstract
Trauma in the adult mammalian central nervous system (CNS) has devastating clinical consequences due to the failure of injured axons to spontaneously regenerate. Over 20 years ago, pioneering work demonstrated that the non-permissive nature of CNS myelin for axon outgrowth contributes to this regenerative failure. Over the past few years, tremendous progress has been made in our understanding of the inhibitory components of CNS myelin, the axonal receptors that respond to these cues, and the intracellular signaling cascades mediating axon outgrowth inhibition. Several approaches designed to antagonize molecular mediators of axon inhibition have been tested in an effort to promote regenerative growth after CNS injury. These studies have validated the role of many candidate proteins in axon outgrowth inhibition; however, other approaches such as the generation of knockout mice for myelin-associated inhibitors have created new questions in the field.
Collapse
Affiliation(s)
- Edith M Grados-Munro
- Montréal Neurological Institute, McGill University Department of Neurology and Neurosurgery, Montréal, Québec, Canada
| | | |
Collapse
|
388
|
Jacobs WB, Fehlings MG. The Molecular Basis of Neural Regeneration. Neurosurgery 2003; 53:943-48; discussion 948-50. [PMID: 14519226 DOI: 10.1227/01.neu.0000083592.74383.b1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2003] [Accepted: 05/07/2003] [Indexed: 11/19/2022] Open
Abstract
THE CENTRAL NERVOUS SYSTEM (CNS) is incapable of meaningful regeneration of lost neurons or axonal and dendritic connections after injury. This often results in permanent and severe loss of neurological function. The CNS regenerative process is unsuccessful for at least three reasons: neurons are highly susceptible to death after CNS injury; the CNS extracellular milieu contains multiple inhibitory factors that make it nonpermissive to growth; and the intrinsic growth capacity of postmitotic neurons is constitutively reduced. However, a number of recent developments in each of these areas is providing insight into the cellular mechanisms involved in CNS regeneration and may eventually lead to the development of therapies capable of effecting successful CNS regeneration.
Collapse
Affiliation(s)
- W Bradley Jacobs
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
389
|
Zhou C, Li Y, Nanda A, Zhang JH. HBO suppresses Nogo-A, Ng-R, or RhoA expression in the cerebral cortex after global ischemia. Biochem Biophys Res Commun 2003; 309:368-76. [PMID: 12951059 DOI: 10.1016/j.bbrc.2003.08.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Nogo-A, a myelin-associated neurite outgrowth inhibitory protein, binds with the Ng-R receptor to activate RhoA intracellular signals and inhibit the plasticity after CNS injury. We evaluated the effect of hyperbaric oxygen (HBO) on the expression of Nogo-A, Ng-R, and RhoA after transient global ischemia in a rat 2 vessel occlusion global ischemic model. Male SD rats (n=78) were randomly divided into 13 groups: 1 sham group, 6 groups of global ischemia, and 6 groups of HBO treatment after global ischemia. HBO (3ATA) was applied for 2 hr at 1 hr after global ischemia. Rats were sacrificed at 6, 12, 24, 48, and 96 hr and 7 days. Global ischemia (10 min) produced a marked increase of Nogo-A/B, Nogo-A, Ng-R, and RhoA expression. Immunohistochemistry showed increased Nogo-A/B and Nogo-A located in the myelin sheath of ischemic brain cortex. Ng-R expressed on the surface of neurons and their processes, and RhoA expressed inside the cytoplasm of neurons in ischemic brain. HBO significantly reduced neurological injury, decreased the levels of Nogo-A, Ng-R, and RhoA in ischemic injured cortex (p<0.05).
Collapse
Affiliation(s)
- Changman Zhou
- Department of Neurosurgery, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | | | | | | |
Collapse
|
390
|
Filbin MT. Myelin-associated inhibitors of axonal regeneration in the adult mammalian CNS. Nat Rev Neurosci 2003; 4:703-13. [PMID: 12951563 DOI: 10.1038/nrn1195] [Citation(s) in RCA: 649] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Marie T Filbin
- Department of Biological Sciences, Hunter College, City University of New York, 695 Park Avenue, New York, New York 10021, USA.
| |
Collapse
|
391
|
Schnaar RL. Myelin molecules limiting nervous system plasticity. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2003; 32:125-42. [PMID: 12827974 DOI: 10.1007/978-3-642-55557-2_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- R L Schnaar
- Departments of Pharmacology and Neuroscience, Johns Hopkins School of Medicine, 725 N. Wolfe Street, Baltimore, Maryland 21205, USA
| |
Collapse
|
392
|
Blixt O, Collins BE, van den Nieuwenhof IM, Crocker PR, Paulson JC. Sialoside specificity of the siglec family assessed using novel multivalent probes: identification of potent inhibitors of myelin-associated glycoprotein. J Biol Chem 2003; 278:31007-19. [PMID: 12773526 DOI: 10.1074/jbc.m304331200] [Citation(s) in RCA: 188] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Ten of the 11 known human siglecs or their murine orthologs have been evaluated for their specificity for over 25 synthetic sialosides representing most of the major sequences terminating carbohydrate groups of glycoproteins and glycolipids. Analysis has been performed using a novel multivalent platform comprising biotinylated sialosides bound to a streptavidin-alkaline phosphatase conjugate. Each siglec was found to have a unique specificity for binding 16 different sialoside-streptavidin-alkaline phosphatase probes. The relative affinities of monovalent sialosides were assessed for each siglec in competitive inhibition studies. The quantitative data obtained allows a detailed analysis of each siglec for the relative importance of sialic acid and the penultimate oligosaccharide sequence on binding affinity and specificity. Most remarkable was the finding that myelin-associated glycoprotein (Siglec-4) binds with 500-10,000-fold higher affinity to a series of mono- and di-sialylated derivatives of the O-linked T-antigen (Galbeta(1-3)-GalNAc(alpha)OThr) as compared with alpha-methyl-NeuAc.
Collapse
MESH Headings
- Alkaline Phosphatase
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- CHO Cells
- Carbohydrate Sequence
- Cell Adhesion Molecules
- Chimera
- Cricetinae
- Enzyme-Linked Immunosorbent Assay
- Galactosides/chemical synthesis
- Galactosides/metabolism
- Glycolipids/chemical synthesis
- Glycolipids/metabolism
- Glycoproteins/chemical synthesis
- Glycoproteins/metabolism
- Humans
- Lectins/genetics
- Lectins/metabolism
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Molecular Sequence Data
- N-Acetylneuraminic Acid/metabolism
- Protein Binding
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Sialic Acid Binding Ig-like Lectin 1
- Sialic Acid Binding Ig-like Lectin 2
- Sialic Acid Binding Immunoglobulin-like Lectins
- Streptavidin
Collapse
Affiliation(s)
- Ola Blixt
- Scripps Research Institute, Department of Molecular Biology, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
393
|
Abstract
Nogo-A is a potent neurite growth inhibitor in vitro and plays a role both in the restriction of axonal regeneration after injury and in structural plasticity in the CNS of higher vertebrates. The regions that mediate inhibition and the topology of the molecule in the plasma membrane have to be defined. Here we demonstrate the presence of three different active sites: (1) an N-terminal region involved in the inhibition of fibroblast spreading, (2) a stretch encoded by the Nogo-A-specific exon that restricts neurite outgrowth and cell spreading and induces growth cone collapse, and (3) a C-terminal region (Nogo-66) with growth cone collapsing function. We show that Nogo-A-specific active fragments bind to the cell surface of responsive cells and to rat brain cortical membranes, suggesting the existence of specific binding partners or receptors. Several antibodies against different epitopes on the Nogo-A-specific part of the protein as well as antisera against the 66 aa loop in the C-terminus stain the cell surface of living cultured oligodendrocytes. Nogo-A is also labeled by nonmembrane-permeable biotin derivatives applied to living oligodendrocyte cultures. Immunofluorescent staining of intracellular, endoplasmic reticulum-associated Nogo-A in cells after selective permeabilization of the plasma membrane reveals that the epitopes of Nogo-A, shown to be accessible at the cell surface, are exposed to the cytoplasm. This suggests that Nogo-A could have a second membrane topology. The two proposed topological variants may have different intracellular as well as extracellular functions.
Collapse
|
394
|
Liu YY, Jin WL, Liu HL, Ju G. Electron microscopic localization of Nogo-A at the postsynaptic active zone of the rat. Neurosci Lett 2003; 346:153-6. [PMID: 12853107 DOI: 10.1016/s0304-3940(03)00508-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Our previous study has demonstrated intraneuronal localization of Nogo-A in motoneurons of the adult rat spinal cord. In the present study, we wished to explore Nogo-A immunoreactivity at synapses using pre-embedding and post-embedding immunogold cytochemistry. Immunogold particles for Nogo-A could be identified in dendrites of the spinal cord motoneurons. Some Nogo-A immunogold particles were found in close apposition to the postsynaptic density and the postsynaptic dense bodies at both symmetric and asymmetric synapses. The present finding of Nogo-A at the postsynaptic active zone provides a morphological basis for the possibility that the Nogo-A-Nogo receptor system may contribute to structural plasticity at synapses as well as along the axonal pathway.
Collapse
Affiliation(s)
- Ying Ying Liu
- Institute of Neurosciences, The Fourth Military Medical University, 17 Chang Le Xi Road, Xi'an, 710032, PR China
| | | | | | | |
Collapse
|
395
|
Hafidi A, Galifianakis D. Macroglia distribution in the developing and adult inferior colliculus. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 143:167-77. [PMID: 12855188 DOI: 10.1016/s0165-3806(03)00110-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Macroglia distribution in the developing and adult gerbil inferior colliculus (IC) was investigated using two oligodendrocytic [myelin-associated-glycoprotein (MAG) and oligodendrocyte-specific molecule (Rip)] and two astrocytic [glial fibrillary acidic protein (GFAP) and S100] markers and immunohistochemistry. There was a spatio-temporal pattern of myelin marker expression starting in the ventral area of the IC and continuing to the dorsal part of the nucleus. Myelination, as revealed by MAG and Rip markers, starts in the IC during the second postnatal week. The intensity of myelination increased between stages P15 and P21 and extended to the whole IC. The appearance of myelin proteins in the IC may suggest a possible axonal outgrowth inhibition by oligodendrocytes in this structure. A differential pattern of staining was obtained with S100 and GFAP antibodies. Astrocytes identified as S100 immunoreactive cells were observed in the IC by birth and the staining was localized to their cell body and processes. S100 positive cells were homogeneously distributed within the IC nucleus. S100 pattern of staining remained the same in stages P7, P15 and P21. In adult IC, S100 positive cell processes were in contact with neuronal cell bodies, other S100 positive cells and blood vessels. Quantitative analysis showed an increase in the density of positive cells during the first postnatal week and a decrease then after through to adulthood. Unlike S100, GFAP immunoreactivity showed a different pattern of staining. At birth GFAP positive astrocytes were observed along the collicular brain midline and around the IC nucleus delimiting its boundaries. The GFAP pattern of labelling remained the same during development and in the adult. This data suggests the presence of two astrocytes subtypes with different locations in the IC nucleus. The GFAP positive astrocytes were located along the edge of the nucleus, while the S100 positive ones displayed a homogeneous distribution across the nucleus.
Collapse
Affiliation(s)
- Aziz Hafidi
- Laboratoire de Biologie Cellulaire et Moleculaire de l'Audition, INSERM EMI 99-27, Université Bordeaux-2, Hôpital Pellegrin, PQR3, 33076 Bordeaux, France.
| | | |
Collapse
|
396
|
Barton WA, Liu BP, Tzvetkova D, Jeffrey PD, Fournier AE, Sah D, Cate R, Strittmatter SM, Nikolov DB. Structure and axon outgrowth inhibitor binding of the Nogo-66 receptor and related proteins. EMBO J 2003; 22:3291-302. [PMID: 12839991 PMCID: PMC165649 DOI: 10.1093/emboj/cdg325] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The myelin-derived proteins Nogo, MAG and OMgp limit axonal regeneration after injury of the spinal cord and brain. These cell-surface proteins signal through multi-subunit neuronal receptors that contain a common ligand-binding glycosylphosphatidylinositol-anchored subunit termed the Nogo-66 receptor (NgR). By deletion analysis, we show that the binding of soluble fragments of Nogo, MAG and NgR to cell-surface NgR requires the entire leucine-rich repeat (LRR) region of NgR, but not other portions of the protein. Despite sharing extensive sequence similarity with NgR, two related proteins, NgR2 and NgR3, which we have identified, do not bind Nogo, MAG, OMgp or NgR. To investigate NgR specificity and multi-ligand binding, we determined the crystal structure of the biologically active ligand-binding soluble ectodomain of NgR. The molecule is banana shaped with elongation and curvature arising from eight LRRs flanked by an N-terminal cap and a small C-terminal subdomain. The NgR structure analysis, as well as a comparison of NgR surface residues not conserved in NgR2 and NgR3, identifies potential protein interaction sites important in the assembly of a functional signaling complex.
Collapse
Affiliation(s)
- William A Barton
- Cellular Biochemistry and Biophysics Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
397
|
Kim JE, Bonilla IE, Qiu D, Strittmatter SM. Nogo-C is sufficient to delay nerve regeneration. Mol Cell Neurosci 2003; 23:451-9. [PMID: 12837628 DOI: 10.1016/s1044-7431(03)00076-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Axonal regeneration succeeds in the peripheral but not central nervous system of adult mammals. Peripheral clearance of myelin coupled with selective CNS expression of axon growth inhibitors, such as Nogo, may account for this reparative disparity. To assess the sufficiency of Nogo for limiting axonal regeneration, we generated transgenic mice expressing Nogo-C in peripheral Schwann cells. Nogo-C includes the panisoform inhibitory Nogo-66 domain, but not a second Nogo-A-specific inhibitory domain, allowing a selective consideration of the Nogo-66 region. The oct-6::nogo-c transgenic mice regenerate axons less rapidly than do wild-type mice after mid-thigh sciatic nerve crush. The delayed axonal regeneration is associated with a decreased recovery rate for motor function after sciatic nerve injury. Thus, expression of the Nogo-66 domain by otherwise permissive myelinating cells is sufficient to hinder axonal reextension after trauma.
Collapse
Affiliation(s)
- Ji Eun Kim
- Departments of Neurology and Neurobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
398
|
Oertle T, Klinger M, Stuermer CAO, Schwab ME. A reticular rhapsody: phylogenic evolution and nomenclature of the RTN/Nogo gene family. FASEB J 2003; 17:1238-47. [PMID: 12832288 DOI: 10.1096/fj.02-1166hyp] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Reticulon (RTN) genes code for a family of proteins relatively recently described in higher vertebrates. The four known mammalian paralogues (RTN1, -2, -3, and -4/Nogo) have homologous carboxyl termini with two characteristic large hydrophobic regions. Except for RTN4-A/Nogo-A, thought to be an inhibitor for neurite outgrowth, restricting the regenerative capabilities of the mammalian CNS after injury, the functions of other family members are largely unknown. The overall occurrence of RTNs in different phyla and the evolution of the RTN gene family have hitherto not been analyzed. Here we expound data showing that the RTN family has arisen during early eukaryotic evolution potentially concerted to the establishment of the endomembrane system. Over 250 reticulon-like (RTNL) genes were identified in deeply diverging eukaryotes, fungi, plants, and animals. A systematic nomenclature for all identified family members is introduced. The analysis of exon-intron arrangements and of protein homologies allowed us to isolate key steps in the history of these genes. Our data corroborate the hypothesis that present RTNs evolved from an intron-rich reticulon ancestor mainly by the loss of different introns in diverse phyla. We also present evidence that the exceptionally large RTN4-A-specific exon 3, which harbors a potent neurite growth inhibitory region, may have arisen de novo approximately 350 MYA during transition to land vertebrates. These data emphasize on the one hand the universal role of reticulons in the eukaryotic system and on the other hand the acquisition of putative new functions through acquirement of novel amino-terminal exons.
Collapse
Affiliation(s)
- Thomas Oertle
- Brain Research Institute, University of Zurich, and Department of Biology, ETH Zurich, 8057 Zurich, Switzerland.
| | | | | | | |
Collapse
|
399
|
Abstract
Traumatized axons possess an extremely limited ability to regenerate within the adult mammalian CNS. The myelin-derived axon outgrowth inhibitors Nogo, oligodendrocyte-myelin glycoprotein, and myelin-associated glycoprotein, all bind to an axonal Nogo-66 receptor (NgR) and at least partially account for this lack of CNS repair. Although the intrathecal application of an NgR competitive antagonist at the time of spinal cord hemisection induces significant regeneration of corticospinal axons, such immediate local therapy may not be as clinically feasible for cases of spinal cord injury. Here, we consider whether this approach can be adapted to systemic therapy in a postinjury therapeutic time window. Subcutaneous treatment with the NgR antagonist peptide NEP1-40 (Nogo extracellular peptide, residues 1-40) results in extensive growth of corticospinal axons, sprouting of serotonergic fibers, upregulation of axonal growth protein SPRR1A (small proline-rich repeat protein 1A), and synapse re-formation. Locomotor recovery after thoracic spinal cord injury is enhanced. Furthermore, delaying the initiation of systemic NEP1-40 administration for up to 1 week after cord lesions does not limit the degree of axon sprouting and functional recovery. This indicates that the regenerative capacity of transected corticospinal tract axons persists for weeks after injury. Systemic Nogo-66 receptor antagonists have therapeutic potential for subacute CNS axonal injuries such as spinal cord trauma.
Collapse
|
400
|
Wang W, Yang Y, Li L, Shi Y. Synleurin, a novel leucine-rich repeat protein that increases the intensity of pleiotropic cytokine responses. Biochem Biophys Res Commun 2003; 305:981-8. [PMID: 12767927 DOI: 10.1016/s0006-291x(03)00876-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have identified and characterized a novel single span transmembrane leucine-rich repeat protein, synleurin, that renders cells highly sensitive to the activation by cytokines and lipopolysaccharide (LPS). The major part of the extracellular domain consists of a leucine-rich repeats (LRR) cassette. The LRR central core has 12 analogous LRR repeating modules arranged in a seamless tandem array. The LRRs are most homologous to that of chondroadherin, insulin-like growth factor binding proteins, platelet glycoprotein V, slits, and toll-like receptors. Synleurin expression was detected at low levels in many tissues, including smooth muscle, brain, uterus, pancreas, cartilage, adipose, spleen, and testis. When synleurin is ecotopically expressed in transfected cells, the cells exhibit amplified responses to bFGF, EGF, PDGF-B, IGF-1, IGF-2, and LPS. Synleurin gene (slrn) maps to human chromosome at 5q12. The name synleurin reflects its synergistic effect on cytokine stimulation and its prominent leucine-rich repeats.
Collapse
Affiliation(s)
- Wei Wang
- Human Genome Sciences, Inc., 9410 Key West Avenue, Rockville, MD 20850, USA
| | | | | | | |
Collapse
|