1
|
Roy A, Pathak Z, Kumar H. Strategies to neutralize RhoA/ROCK pathway after spinal cord injury. Exp Neurol 2021; 343:113794. [PMID: 34166685 DOI: 10.1016/j.expneurol.2021.113794] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/01/2021] [Accepted: 06/19/2021] [Indexed: 01/22/2023]
Abstract
Regeneration is bungled following CNS injuries, including spinal cord injury (SCI). Inherent decay of permissive conditions restricts the regrowth of the mature CNS after an injury. Hypertrophic scarring, insignificant intrinsic axon-growth activity, and axon-growth inhibitory molecules such as myelin inhibitors and scar inhibitors constitute a significant hindrance to spinal cord repair. Besides these molecules, a combined absence of various mechanisms responsible for axonal regeneration is the main reason behind the dereliction of the adult CNS to regenerate. The neutralization of specific inhibitors/proteins by stymieing antibodies or encouraging enzymatic degradation results in improved axon regeneration. Previous efforts to induce regeneration after SCI have stimulated axonal development in or near lesion sites, but not beyond them. Several pathways are responsible for the axonal growth obstruction after a CNS injury, including SCI. Herein, we summarize the axonal, glial, and intrinsic factor which impedes the regeneration. We have also discussed the methods to stabilize microtubules and through this to maintain the proper cytoskeletal dynamics of growth cone as disorganized microtubules lead to the failure of axonal regeneration. Moreover, we primarily focus on diverse inhibitors of axonal growth and molecular approaches to counteract them and their downstream intracellular signaling through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Zarna Pathak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
2
|
Wu D, Jin Y, Shapiro TM, Hinduja A, Baas PW, Tom VJ. Chronic neuronal activation increases dynamic microtubules to enhance functional axon regeneration after dorsal root crush injury. Nat Commun 2020; 11:6131. [PMID: 33257677 PMCID: PMC7705672 DOI: 10.1038/s41467-020-19914-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022] Open
Abstract
After a dorsal root crush injury, centrally-projecting sensory axons fail to regenerate across the dorsal root entry zone (DREZ) to extend into the spinal cord. We find that chemogenetic activation of adult dorsal root ganglion (DRG) neurons improves axon growth on an in vitro model of the inhibitory environment after injury. Moreover, repeated bouts of daily chemogenetic activation of adult DRG neurons for 12 weeks post-crush in vivo enhances axon regeneration across a chondroitinase-digested DREZ into spinal gray matter, where the regenerating axons form functional synapses and mediate behavioral recovery in a sensorimotor task. Neuronal activation-mediated axon extension is dependent upon changes in the status of tubulin post-translational modifications indicative of highly dynamic microtubules (as opposed to stable microtubules) within the distal axon, illuminating a novel mechanism underlying stimulation-mediated axon growth. We have identified an effective combinatory strategy to promote functionally-relevant axon regeneration of adult neurons into the CNS after injury.
Collapse
Affiliation(s)
- Di Wu
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Ying Jin
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Tatiana M Shapiro
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Abhishek Hinduja
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Peter W Baas
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Yu Y, Wang D, Li H, Liu Y, Xiang Z, Wu J, Jing X. IPSC‑MSC inhibition assessment in Raw 264.7 cells following oxygen and glucose deprivation reveals a distinct function for cardiopulmonary resuscitation. Mol Med Rep 2018; 17:8212-8220. [PMID: 29658608 PMCID: PMC5983996 DOI: 10.3892/mmr.2018.8864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 11/02/2017] [Indexed: 01/01/2023] Open
Abstract
Hypoxia is a serious stress state. The nervous system is less tolerant to hypoxia, and cell death due to hypoxia is irreversible. With the incidence of cardiovascular disease gradually increasing, the sudden cardiac death rate is additionally increasing. Although cardiopulmonary resuscitation (CPR) is an important development, recovery is frequently poor. In a successful recovery population, ~40% of the population was in a vegetative state or subsequently succumbed to their condition, and ~20% had brain damage. Therefore, the recovery of the brain is of particular importance in CPR. Immune disorders are one of the major mechanisms of cerebral resuscitation following CPR. Studies have demonstrated that induced pluripotent stem cell-derived mesenchymal stem cells (IPSC-MSCs) have a strong immune regulatory effect during tissue repair and anti-inflammatory effects. IPSC-MSCs may inhibit the inflammatory response by means of the inflammatory reaction network to improve brain function following CPR, although the cellular and molecular mechanisms remain unclear. Macrophages are a bridge between innate immune and specific immune responses in the body; therefore, it was hypothesized that macrophages may be the important effector cell of the role of IPSC-MSCs in improving brain function following recovery of spontaneous respiration and circulation subsequent to cardiopulmonary resuscitation. In the present study, IPSC-MSCs were applied to the oxygen and glucose deprivation (OGD) model. It was observed that intervention with IPSC-MSCs was able to alter the polarization direction of macrophages. The difference in the proportions of M1 and M2 macrophages was statistically significant at 6, 12, 24 and 48 h (P=0.037, P<0.05) in the OGD + IPSC-MSCs group (M1, 33.48±5.6%; M2, 50.84±6.9%) and in the OGD group (M1, 83.55±7.3%; M2, 11.41±3.2%), and over time this trend was more obvious. The polarization direction of macrophages is associated with the neurogenic locus notch homolog protein 1 (Notch-1) signaling pathway. In conclusion, it was observed that IPSC-MSCs may be associated with altered macrophage polarization, which may be accomplished by inhibiting the Notch-1 signaling pathway.
Collapse
Affiliation(s)
- Yi Yu
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Dongping Wang
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hui Li
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yujie Liu
- Department of Breast Surgery and Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat‑Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhao Xiang
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Junlin Wu
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaoli Jing
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
4
|
Wu D, Klaw MC, Kholodilov N, Burke RE, Detloff MR, Côté MP, Tom VJ. Expressing Constitutively Active Rheb in Adult Dorsal Root Ganglion Neurons Enhances the Integration of Sensory Axons that Regenerate Across a Chondroitinase-Treated Dorsal Root Entry Zone Following Dorsal Root Crush. Front Mol Neurosci 2016; 9:49. [PMID: 27458339 PMCID: PMC4932115 DOI: 10.3389/fnmol.2016.00049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/07/2016] [Indexed: 11/13/2022] Open
Abstract
While the peripheral branch of dorsal root ganglion neurons (DRG) can successfully regenerate after injury, lesioned central branch axons fail to regrow across the dorsal root entry zone (DREZ), the interface between the dorsal root and the spinal cord. This lack of regeneration is due to the limited regenerative capacity of adult sensory axons and the growth-inhibitory environment at the DREZ, which is similar to that found in the glial scar after a central nervous system (CNS) injury. We hypothesized that transduction of adult DRG neurons using adeno-associated virus (AAV) to express a constitutively-active form of the GTPase Rheb (caRheb) will increase their intrinsic growth potential after a dorsal root crush. Additionally, we posited that if we combined that approach with digestion of upregulated chondroitin sulfate proteoglycans (CSPG) at the DREZ with chondroitinase ABC (ChABC), we would promote regeneration of sensory axons across the DREZ into the spinal cord. We first assessed if this strategy promotes neuritic growth in an in vitro model of the glial scar containing CSPG. ChABC allowed for some regeneration across the once potently inhibitory substrate. Combining ChABC treatment with expression of caRheb in DRG significantly improved this growth. We then determined if this combination strategy also enhanced regeneration through the DREZ after dorsal root crush in adult rats in vivo. After unilaterally crushing C4-T1 dorsal roots, we injected AAV5-caRheb or AAV5-GFP into the ipsilateral C5-C8 DRGs. ChABC or PBS was injected into the ipsilateral dorsal horn at C5-C8 to digest CSPG, for a total of four animal groups (caRheb + ChABC, caRheb + PBS, GFP + ChABC, GFP + PBS). Regeneration was rarely observed in PBS-treated animals, whereas short-distance regrowth across the DREZ was observed in ChABC-treated animals. No difference in axon number or length between the ChABC groups was observed, which may be related to intraganglionic inflammation induced by the injection. ChABC-mediated regeneration is functional, as stimulation of ipsilateral median and ulnar nerves induced neuronal c-Fos expression in deafferented dorsal horn in both ChABC groups. Interestingly, caRheb + ChABC animals had significantly more c-Fos+ nuclei indicating that caRheb expression in DRGs promoted functional synaptogenesis of their axons that regenerated beyond a ChABC-treated DREZ.
Collapse
Affiliation(s)
- Di Wu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| | - Michelle C Klaw
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| | - Nikolai Kholodilov
- Department of Neurology, Columbia University in the City of New York New York, NY, USA
| | - Robert E Burke
- Department of Neurology, Columbia University in the City of New YorkNew York, NY, USA; Department of Pathology and Cell Biology, Columbia University in the City of New YorkNew York, NY, USA
| | - Megan R Detloff
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| | - Marie-Pascale Côté
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| |
Collapse
|
5
|
Silva NA, Sousa N, Reis RL, Salgado AJ. From basics to clinical: a comprehensive review on spinal cord injury. Prog Neurobiol 2013; 114:25-57. [PMID: 24269804 DOI: 10.1016/j.pneurobio.2013.11.002] [Citation(s) in RCA: 555] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 11/12/2013] [Accepted: 11/12/2013] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological disorder that affects thousands of individuals each year. Over the past decades an enormous progress has been made in our understanding of the molecular and cellular events generated by SCI, providing insights into crucial mechanisms that contribute to tissue damage and regenerative failure of injured neurons. Current treatment options for SCI include the use of high dose methylprednisolone, surgical interventions to stabilize and decompress the spinal cord, and rehabilitative care. Nonetheless, SCI is still a harmful condition for which there is yet no cure. Cellular, molecular, rehabilitative training and combinatorial therapies have shown promising results in animal models. Nevertheless, work remains to be done to ascertain whether any of these therapies can safely improve patient's condition after human SCI. This review provides an extensive overview of SCI research, as well as its clinical component. It starts covering areas from physiology and anatomy of the spinal cord, neuropathology of the SCI, current clinical options, neuronal plasticity after SCI, animal models and techniques to assess recovery, focusing the subsequent discussion on a variety of promising neuroprotective, cell-based and combinatorial therapeutic approaches that have recently moved, or are close, to clinical testing.
Collapse
Affiliation(s)
- Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Caldas das Taipas, Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
6
|
Pandey S, Alcaro MC, Scrima M, Peroni E, Paolini I, Di Marino S, Barbetti F, Carotenuto A, Novellino E, Papini AM, D'Ursi AM, Rovero P. Designed glucopeptides mimetics of myelin protein epitopes as synthetic probes for the detection of autoantibodies, biomarkers of multiple sclerosis. J Med Chem 2012; 55:10437-47. [PMID: 23167575 DOI: 10.1021/jm301031r] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We previously reported that CSF114(Glc) detects diagnostic autoantibodies in multiple sclerosis sera. We report herein a bioinformatic analysis of myelin proteins and CSF114(Glc), which led to the identification of five sequences. These glucopeptides were synthesized and tested in enzymatic assays, showing a common minimal epitope. Starting from that, we designed an optimized sequence, SP077, showing a higher homology with both CSF114(Glc) and the five sequences selected using the bioinformatic approach. SP077 was synthesized and tested on 50 multiple sclerosis patients' sera, and was able to detect higher antibody titers as compared to CSF114(Glc). Finally, the conformational properties of SP077 were studied by NMR spectroscopy and structure calculations. Thus, the immunological activity of SP077 in the recognition of specific autoantibodies in multiple sclerosis patients' sera may be ascribed to both the optimized design of its epitopic region and the superior surface interacting properties of its C-terminal region.
Collapse
Affiliation(s)
- Shashank Pandey
- Laboratory of Peptide and Protein Chemistry and Biology, Department of Chemistry Ugo Schiff, University of Florence, I-50019 Sesto Fiorentino, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Pathi SS, Jose S, Govindaraju S, Conde JA, Romo HE, Chamakura KR, Claunch CJ, Benito-Martín A, Challa-Malladi M, González-García M, Ballestero RP. zRICH, a protein induced during optic nerve regeneration in zebrafish, promotes neuritogenesis and interacts with tubulin. Brain Res 2012; 1474:29-39. [PMID: 22885342 DOI: 10.1016/j.brainres.2012.07.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 07/07/2012] [Accepted: 07/30/2012] [Indexed: 10/28/2022]
Abstract
Mammals do not regenerate axons in their central nervous system (CNS) spontaneously. This phenomenon is the cause of numerous medical conditions after damage to nerve fibers in the CNS of humans. The study of the mechanisms of nerve regeneration in other vertebrate animals able to spontaneously regenerate axons in their CNS is essential for understanding nerve regeneration from a scientific point of view, and for developing therapeutic approaches to enhance nerve regeneration in the CNS of humans. RICH proteins are a novel group of proteins implicated in nerve regeneration in the CNS of teleost fish, yet their mechanisms of action are not well understood. A number of mutant versions of the zebrafish RICH (zRICH) protein were generated and characterized at biochemical and cellular levels in our laboratory. With the aim of understanding the effects of RICH proteins in neuronal axon outgrowth, stable transfectants derived from the neuronal model PC12 cell line expressing zRICH Wild-Type or mutant versions of zRICH were studied. Results from differentiation experiments suggest that RICH proteins enhance neuronal plasticity by facilitating neurite branching. Biochemical co-purification results have demonstrated that zRICH binds to the cytoskeletal protein tubulin. The central domain of the protein is sufficient for tubulin binding, but a mutant version of the protein lacking the terminal domains, which cannot bind to the plasma membrane, was not able to enhance neurite branching. RICH proteins may facilitate axon regeneration by regulating the axonal cytoskeleton and facilitating the formation of new neurite branches.
Collapse
Affiliation(s)
- Satya S Pathi
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Sun Y, Lim Y, Li F, Liu S, Lu JJ, Haberberger R, Zhong JH, Zhou XF. ProBDNF collapses neurite outgrowth of primary neurons by activating RhoA. PLoS One 2012; 7:e35883. [PMID: 22558255 PMCID: PMC3338794 DOI: 10.1371/journal.pone.0035883] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 03/23/2012] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Neurons extend their dendrites and axons to build functional neural circuits, which are regulated by both positive and negative signals during development. Brain-derived neurotrophic factor (BDNF) is a positive regulator for neurite outgrowth and neuronal survival but the functions of its precursor (proBDNF) are less characterized. METHODOLOGY/PRINCIPAL FINDINGS Here we show that proBDNF collapses neurite outgrowth in murine dorsal root ganglion (DRG) neurons and cortical neurons by activating RhoA via the p75 neurotrophin receptor (p75NTR). We demonstrated that the receptor proteins for proBDNF, p75NTR and sortilin, were highly expressed in cultured DRG or cortical neurons. ProBDNF caused a dramatic neurite collapse in a dose-dependent manner and this effect was about 500 fold more potent than myelin-associated glycoprotein. Neutralization of endogenous proBDNF by using antibodies enhanced neurite outgrowth in vitro and in vivo, but this effect was lost in p75NTR(-/-) mice. The neurite outgrowth of cortical neurons from p75NTR deficient (p75NTR(-/-)) mice was insensitive to proBDNF. There was a time-dependent reduction of length and number of filopodia in response to proBDNF which was accompanied with a polarized RhoA activation in growth cones. Moreover, proBDNF treatment of cortical neurons resulted in a time-dependent activation of RhoA but not Cdc42 and the effect was absent in p75NTR(-/-) neurons. Rho kinase (ROCK) and the collapsin response mediator protein-2 (CRMP-2) were also involved in the proBDNF action. CONCLUSIONS proBDNF has an opposing role in neurite outgrowth to that of mature BDNF. Our observations suggest that proBDNF collapses neurites outgrowth and filopodial growth cones by activating RhoA through the p75NTR signaling pathway.
Collapse
MESH Headings
- Adaptor Proteins, Vesicular Transport/genetics
- Adaptor Proteins, Vesicular Transport/metabolism
- Animals
- Antibodies/pharmacology
- Brain-Derived Neurotrophic Factor/antagonists & inhibitors
- Brain-Derived Neurotrophic Factor/pharmacology
- Brain-Derived Neurotrophic Factor/physiology
- Cells, Cultured
- Ganglia, Spinal/cytology
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/physiology
- Gene Expression Regulation, Developmental/physiology
- Mice
- Mice, Knockout
- Nerve Fibers/drug effects
- Nerve Fibers/physiology
- Neurites/drug effects
- Neurites/physiology
- Protein Precursors/pharmacology
- Protein Precursors/physiology
- Pseudopodia/drug effects
- Pseudopodia/physiology
- Receptors, Nerve Growth Factor/deficiency
- Receptors, Nerve Growth Factor/genetics
- Signal Transduction/physiology
- Time-Lapse Imaging
- rho GTP-Binding Proteins/agonists
- rho GTP-Binding Proteins/genetics
- rho GTP-Binding Proteins/metabolism
- rhoA GTP-Binding Protein
Collapse
Affiliation(s)
- Ying Sun
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, Australia
| | - Yoon Lim
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, Australia
- Division of Health Science, Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Fang Li
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, Australia
| | - Shen Liu
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, Australia
- Division of Health Science, Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Jian-Jun Lu
- Division of Health Science, Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Rainer Haberberger
- Department of Anatomy and Histology and Centre for Neuroscience, Flinders University, Adelaide, Australia
| | - Jin-Hua Zhong
- Division of Health Science, Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Xin-Fu Zhou
- Division of Health Science, Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
9
|
Tan HB, Zhong YS, Cheng Y, Shen X. Rho/ROCK pathway and neural regeneration: a potential therapeutic target for central nervous system and optic nerve damage. Int J Ophthalmol 2011; 4:652-7. [PMID: 22553739 DOI: 10.3980/j.issn.2222-3959.2011.06.16] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 11/29/2011] [Indexed: 01/12/2023] Open
Abstract
Rho-associated kinase (ROCK) is a serine/threonine kinase and one of the major downstream effectors of the small GTPase RhoA. The Rho/ROCK pathway is closely related to the pathogenesis of several central nervous system (CNS) disorders, and involved in many aspects of neuronal functions including neurite outgrowth and retraction. In the adult CNS, the damaged neuron regeneration is very difficult due to the presence of myelin-associated axon growth inhibitors such as Nogo, myelin-associated glycoprotein (MAG) and oligodendrocyte-myelin glycoprotein (Omgp), etc. The effects of these axon growth inhibitors are reversed by blocking the Rho/ROCK pathway in vitro, and the inhibition of Rho/ROCK pathway can promote axon regeneration and functional recovery in the injured CNS in vivo. In addition, the therapeutic effects of the Rho/ROCK inhibitors have also been demonstrated in some animal models and the Rho/ROCK pathway becomes an attractive target for the development of drugs for treating CNS disorders. In this review, we summarized on the effect of the Rho and the downstream factor ROCK in neural regeneration, and the potential therapeutic effect of Rho/ROCK inhibitors in the survival and axonal regeneration of retinal ganglion cells was also discussed.
Collapse
Affiliation(s)
- Hai-Bo Tan
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, Shanghai 200025, China
| | | | | | | |
Collapse
|
10
|
Lv B, Yuan W, Xu S, Zhang T, Liu B. Lentivirus-siNgR199 Promotes Axonal Regeneration and Functional Recovery in Rats. Int J Neurosci 2011; 122:133-9. [DOI: 10.3109/00207454.2011.633720] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
11
|
Riboldi G, Nizzardo M, Simone C, Falcone M, Bresolin N, Comi GP, Corti S. ALS genetic modifiers that increase survival of SOD1 mice and are suitable for therapeutic development. Prog Neurobiol 2011; 95:133-48. [PMID: 21816207 DOI: 10.1016/j.pneurobio.2011.07.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 07/19/2011] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a frequently fatal motor neuron disease without any cure. To find molecular therapeutic targets, several studies crossed transgenic ALS murine models with animals transgenic for some ALS target genes. We aimed to revise the new discoveries and new works in this field. We selected the 10 most promising genes, according to their capability when down-regulated or up-regulated in ALS animal models, for increasing life span and mitigating disease progression: XBP-1, NogoA and NogoB, dynein, heavy and medium neurofilament, NOX1 and NOX2, MLC-mIGF-1, NSE-VEGF, and MMP-9. Interestingly, some crucial modifier genes have been described as being involved in common pathways, the most significant of which are inflammation and cytoskeletal activities. The endoplasmic reticulum also seems to play an important role in ALS pathogenesis, as it is involved in different selected gene pathways. In addition, these genes have evident links to each other, introducing the hypothesis of a single unknown, common pathway involving all of these identified genes and others to be discovered.
Collapse
Affiliation(s)
- Giulietta Riboldi
- Department of Neurological Sciences, Dino Ferrari Centre, University of Milan, IRCCS Fondazione Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
12
|
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
|
13
|
Chen C, Chen X, Yin X, Yuan R, Wang B, Ye J. NgR RNA interference, combined with zymosan intravitreal injection, enhances optic nerve regeneration. J Neurochem 2009; 110:1628-34. [PMID: 19575706 DOI: 10.1111/j.1471-4159.2009.06264.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mature retinal ganglion cells like other CNS neurons are unable to regenerate their axons after injury. Regenerative failure has been attributed, in part, to two factors: the existence of myelin-derived inhibitors that bind to the Nogo receptor (NgR) and a deficiency of trophic support factors. We investigated the regrowth of injured axons both by inhibiting NgR by RNA interference and by recruiting exogenous trophic support by zymosan intravitreal injection. Our results showed that either approach can stimulate optic nerve axon regrowth but regenerated axons can grow longer and extend further when both methods are combined. We conclude that endogenous NgR inhibition and exogenous trophic support both play independent, important roles in enhancing optic nerve axon regrowth and that the regenerative effect can be augmented when the two are combined. This may provide a therapeutic strategy for promoting axon regeneration in the CNS as well.
Collapse
Affiliation(s)
- Chunlin Chen
- Department of Ophthalmology, Research Institute of Field Surgery, Da Ping Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
14
|
Jiang W, Xia F, Han J, Wang J. Patterns of Nogo-A, NgR, and RhoA expression in the brain tissues of rats with focal cerebral infarction. Transl Res 2009; 154:40-8. [PMID: 19524873 DOI: 10.1016/j.trsl.2009.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 04/10/2009] [Accepted: 04/14/2009] [Indexed: 01/08/2023]
Abstract
Nogo-A and its Nogo receptor (NgR) have been shown to inhibit plasticity after central nervous system lesions. Therefore, we hypothesized that Nogo-A and its receptor NgR will be upregulated and will activate RhoA, and thus, they play a role in the damage in the infarction developed. To test this hypothesis, a focal cerebral infarction model was created by coagulation of the right middle cerebral artery (MCA) and ipsilateral common carotid artery (CCA), as well as the simultaneous transient occlusion of the contralateral CCA for 30 min in 60 adult Sprague-Dawley rats. The rat brains were treated at 6 h, 12 h, 24 h, 48 h, 96 h, and 7 d after cerebral infarction. Sham controls were collected to determine histopathologic damage and Nogo-A, NgR, and RhoA expression using hematoxylin-eosin, immunohistochemical staining, Western blot analysis, and fluorimeter-based quantitive reverse transcriptase-polymerase chain reaction. The results indicate that cerebral infarction produced damage and edema on nerve cells in the infarction area, becoming most prominent at 24h after modeling. Meanwhile, a marked increase of Nogo-A, NgR, and RhoA expression was found at 6h in model groups compared with the sham controls, which peaked at 24 h after the operation. Immunohistochemical staining and Western blot analysis also showed upregulated Nogo-A located in the myelin sheath of the infarction area, NgR expressed on the surface of neurons and their processes, and RhoA expressed inside the cytoplasm of neurons in infarction brain. In conclusion, the upregulation of Nogo-A, NgR, and RhoA in the infarction area may be an important feature of cerebral infarction and may play a role in the pathologic progression of this lesion.
Collapse
Affiliation(s)
- Wen Jiang
- Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | |
Collapse
|
15
|
Persistent restoration of sensory function by immediate or delayed systemic artemin after dorsal root injury. Nat Neurosci 2008; 11:488-96. [PMID: 18344995 DOI: 10.1038/nn2069] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 02/11/2008] [Indexed: 11/08/2022]
Abstract
Dorsal root injury results in substantial and often irreversible loss of sensory functions as a result of the limited regenerative capacity of sensory axons and the inhibitory barriers that prevent both axonal entry into and regeneration in the spinal cord. Here, we describe previously unknown effects of the growth factor artemin after crush injury of the dorsal spinal nerve roots in rats. Artemin not only promoted re-entry of multiple classes of sensory fibers into the spinal cord and re-establishment of synaptic function and simple behavior, but it also, surprisingly, promoted the recovery of complex behavior. These effects occurred after a 2-week schedule of intermittent, systemic administration of artemin and persisted for at least 6 months following treatment, suggesting a substantial translational advantage. Systemic artemin administration produced essentially complete and persistent restoration of nociceptive and sensorimotor functions, and could represent a promising therapy that may effectively promote sensory neuronal regeneration and functional recovery after injury.
Collapse
|
16
|
Gross RE, Mei Q, Gutekunst CA, Torre E. The pivotal role of RhoA GTPase in the molecular signaling of axon growth inhibition after CNS injury and targeted therapeutic strategies. Cell Transplant 2007; 16:245-62. [PMID: 17503736 DOI: 10.3727/000000007783464740] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The dogma that the adult central nervous system (CNS) is nonpermissive to axonal regeneration is beginning to fall in the face of increased understanding of the molecular and cellular biology of axon outgrowth. It is now appreciated that axon growth is regulated by a combination of extracellular factors related to the milieu of the developing or adult CNS and the presence of injury, and intracellular factors related to the "growth state" of the developing or regenerating neuron. Several critical points of convergence within the developing or regenerating neuron for mediating intracellular cell signaling effects on the growth cone cytoskeleton have been identified, and their modulation has produced marked increases in axon outgrowth within the "nonpermissive" milieu of the adult injured CNS. One such critical convergence point is the small GTPase RhoA, which integrates signaling events produced by both myelin-associated inhibitors (e.g., NogoA) and astroglial-derived inhibitors (chondroitin sulfate proteoglycans) and regulates the activity of downstream effectors that modulate cytoskeletal dynamics within the growth cone mediating axon outgrowth or retraction. Inhibition of RhoA has been associated with increased outgrowth on nonpermissive substrates in vitro and increased axon regeneration in vivo. We are developing lentiviral vectors that modulate RhoA activity, allowing more long-term expression than is possible with current approaches. These vectors may be useful in regenerative strategies for spinal cord injury, brain injury, and neurodegenerative diseases including Parkinson's disease, Alzheimer's disease, and Huntington's disease.
Collapse
Affiliation(s)
- Robert E Gross
- Department of Neurosurgery, Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | |
Collapse
|
17
|
Kawashima R, Kojima H, Nakamura K, Arahata A, Fujita Y, Tokuyama Y, Saito T, Furudate SI, Kurihara T, Yagishita S, Kitamura K, Tamai Y. Alterations in mRNA expression of myelin proteins in the sciatic nerves and brains of streptozotocin-induced diabetic rats. Neurochem Res 2007; 32:1002-10. [PMID: 17404843 DOI: 10.1007/s11064-006-9260-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Accepted: 12/19/2006] [Indexed: 01/06/2023]
Abstract
Diabetic neuropathy is the most common complication of diabetes. We examined the levels and the mRNA expression of myelin proteins in the sciatic nerves and the brains of streptozotocin-induced diabetic rats. The diabetic rats exhibited a decrease in body weight, elevation of the blood glucose level and a decrease in motor nerve conduction velocity at 2 weeks after streptozotocin injection. In the sciatic nerves of diabetic rats, the level of P0 protein and its mRNA expression were markedly reduced at 20 weeks after the injection. In the brains, the levels of proteolipid protein and myelin-associated glycoprotein and their mRNA expression were selectively decreased at 20 weeks after the injection. This affected expression of myelin proteins was found even when no histological abnormalities were detectable. Considering the functional significance of myelin proteins, this impairment of protein expression is possibly involved in the pathogenesis of diabetic neuropathy, including that in brain disorders.
Collapse
Affiliation(s)
- Rei Kawashima
- Department of Biochemistry, Kitasato University School of Medicine, 1-15-1 Kitasato, Sagamihara-shi, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Barrette B, Vallières N, Dubé M, Lacroix S. Expression profile of receptors for myelin-associated inhibitors of axonal regeneration in the intact and injured mouse central nervous system. Mol Cell Neurosci 2007; 34:519-38. [PMID: 17234430 DOI: 10.1016/j.mcn.2006.12.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 11/30/2006] [Accepted: 12/08/2006] [Indexed: 01/12/2023] Open
Abstract
Although CNS neurons have the potential to regenerate their axons after injury, myelin debris carrying axon growth inhibitors rapidly induce growth cone collapse. Receptors (NgR1, NgR2) and coreceptors (LINGO-1, p75(NTR), TROY) for these inhibitors have been characterized and transduction pathways partially identified. However, little is known about the expression of these receptors in intact and lesioned supraspinal projection neurons. Using in situ hybridization, immunohistochemistry and neuronal tract-tracing, we found that NgR1, NgR2 and LINGO-1 are strongly expressed in several neuronal populations of the adult mouse brain projecting to the spinal cord, including neurons projecting through the corticospinal, rubrospinal, caerulospinal, reticulospinal, raphespinal and vestibulospinal tracts. As expected, p75(NTR) expression was restricted to neuronal descending pathways from the brainstem. TROY was absent from most brain regions and from all neuronal projection systems, suggesting that additional signal-transducing coreceptors exist. Qualitative and quantitative analyses revealed that brain expression for these receptors was not affected by a severe T10 spinal cord contusion.
Collapse
Affiliation(s)
- Benoit Barrette
- Department of Anatomy and Physiology, Laval University, Ste-Foy, Québec, Canada
| | | | | | | |
Collapse
|
19
|
Zander H, Reineke U, Schneider-Mergener J, Skerra A. Epitope mapping of the neuronal growth inhibitor Nogo-A for the Nogo receptor and the cognate monoclonal antibody IN-1 by means of the SPOT technique. J Mol Recognit 2007; 20:185-96. [PMID: 17486692 DOI: 10.1002/jmr.823] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Nogo-A is a potent inhibitor of axonal outgrowth in the central nervous system of adult mammals, where it is expressed as a membrane protein on oligodendrocytes and in myelin. Here we describe an attempt to identify linear peptide epitopes in its sequence that are responsible for the interaction either with the Nogo receptor (NgR) or with the neutralizing monoclonal antibody IN-1. Analysis of an array of immobilized overlapping 15 mer peptides covering the entire amino acid sequence of human Nogo-A (1192 residues) revealed a single epitope with prominent binding activity both towards the recombinant NgR and the IN-1 F(ab) fragment. Further truncation and substitution analysis yielded the minimal epitope sequence 'IKxLRRL' (x not equal to P), which occurs within the so-called Nogo66 region (residues 1054-1120) of Nogo-A. The bacterially produced Nogo66 fragment exhibited binding activity both for the recombinant NgR and for the IN-1 F(ab) fragment on the Western blot as well as in ELISA. Unexpectedly, the synthetic epitope peptide and the recombinant Nogo66 showed cross-reactivity with the 8-18C5 F(ab) fragment, which is directed against myelin oligodendrocyte glycoprotein (MOG) as a structurally unrelated target. On the other hand, the recombinant N-terminal domain of Nogo-A (residues 334-966) was shown to specifically interact on the Western blot and in an ELISA with the IN-1 F(ab) fragment but not with the recombinant NgR, which is in agreement with previous results. Hence, our data suggest that there is a distinct binding site for the Nogo receptor in the Nogo66 region of Nogo-A, whereas its interaction with NgR is less specific than anticipated before. Although there probably exists a non-linear epitope for the neutralizing antibody IN-1 in the N-terminal region of Nogo-A, which is likely to be accessible from outside the cell, a previously postulated second binding site for NgR in this region (called Nogo-A-24) remains elusive.
Collapse
Affiliation(s)
- Hilke Zander
- Lehrstuhl für Biologische Chemie, Technische Universität München, An der Saatzucht 5, 85350 Freising-Weihenstephan, Germany
| | | | | | | |
Collapse
|
20
|
Zhu HY, Guo HF, Hou HL, Liu YJ, Sheng SL, Zhou JN. Increased expression of the Nogo receptor in the hippocampus and its relation to the neuropathology in Alzheimer's disease. Hum Pathol 2006; 38:426-34. [PMID: 17188332 DOI: 10.1016/j.humpath.2006.09.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Revised: 08/31/2006] [Accepted: 09/06/2006] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia in human beings. Its best-known pathologic feature is the presence of senile plaques and neurofibrillary tangles in the brain. Nogo-66 receptor (NgR) is believed to contribute to the inhibitory activities of axon regeneration after injury. This study investigated the expression of NgR in the hippocampus and its relation to the pathologic changes of AD using immunohistochemistry and double-labeling immunofluorescence methods. The results showed that NgR immunoreactivity was present in more than 50% of the pyramidal layer cells of the CA1 to CA4 subfields of the hippocampus. No significant difference was observed in the number of NgR immunopositive cells in the CA1 to CA4 subfields between patients with AD and control subjects, whereas the ratio of NgR immunopositive cells to the total number of pyramidal layer cells was revealed to be significantly higher in the CA1 and CA2 subfields of the hippocampus of patients with AD than that in the same region of the control subjects. Moreover, high numbers of AT-8 immunopositive cells were found to be double-labeled with NgR in the CA1 subfields of patients with AD, whereas only few NgR deposits were observed in the senile plaques of the hippocampus in these patients. These results suggest that NgR may be related to the formation of tangles in AD.
Collapse
Affiliation(s)
- Hong-Yan Zhu
- Hefei National Laboratory for Physical Science at the Microscale, University of Science and Technology of China, Hefei 230026, PR China
| | | | | | | | | | | |
Collapse
|
21
|
Challa M, Chapa GR, Govindaraju S, González-García M, Ballestero RP. Characterization of the domains of zRICH, a protein induced during optic nerve regeneration in zebrafish. Brain Res 2006; 1100:42-54. [PMID: 16765331 DOI: 10.1016/j.brainres.2006.04.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Revised: 04/26/2006] [Accepted: 04/30/2006] [Indexed: 01/03/2023]
Abstract
Teleost fish show a remarkable capability of nerve regeneration in their CNS, while injuries to axon fibers in the CNS of mammals result in degeneration and loss of function. Understanding this difference has biomedical consequences to humans. Both extrinsic factors from the neuronal environment and intrinsic neuronal factors seem to play a role in successful nerve regeneration. Among the intrinsic factors, a number of proteins termed axonal growth associated proteins (GAPs) are strongly induced during axon regeneration. RICH proteins are axonal GAPs that show homology to mammalian myelin marker proteins termed CNPases. Sequence analysis distinguishes three domains in these proteins. In this report, mutant versions of zebrafish RICH proteins were generated to study the roles of the domains of the protein at biochemical and cellular levels. The central CNPase homology domain was sufficient for catalytic activity. The amino terminal acidic domain causes the anomalous electrophoretic migration observed for RICH proteins. The small C-terminal domain bears an isoprenylation motif and is necessary for the interaction of zRICH with cellular membranes. At the cellular level, expression of wild-type zRICH protein in PC12 cells did not induce neurite generation. Additionally, neither the expression of wild-type zRICH nor the expression of mutant versions of the protein interfered with the levels of differentiation of PC12 cells induced by nerve growth factor, suggesting that, at least in this model of neuronal differentiation, zRICH proteins do not participate in the process of generation of neurites.
Collapse
Affiliation(s)
- Madhavi Challa
- Department of Chemistry, Texas A&M University-Kingsville, 700 University Boulevard, Kingsville, TX, USA
| | | | | | | | | |
Collapse
|
22
|
Schwab JM, Tuli SK, Failli V. The Nogo receptor complex: confining molecules to molecular mechanisms. Trends Mol Med 2006; 12:293-7. [PMID: 16723274 DOI: 10.1016/j.molmed.2006.05.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Revised: 04/04/2006] [Accepted: 05/11/2006] [Indexed: 01/25/2023]
Abstract
Myelin inhibitory ligands of the Nogo-66 receptor (NgR1) limit axon regeneration in the adult CNS. Recent findings have identified additional co-receptors (functional homologues) of the trimeric NgR1 complex, post-translational modifications of the co-receptors within the cell membrane and novel Ca(2+)-dependent cytoplasmic-protein phosphorylation mechanisms. Such unique signalling pathways provide the potential to transduce myelin-derived growth inhibitory signals to the axonal cytoskeleton, and have been areas of intense investigation in recent years. Here, we summarize current understanding of the molecular basis of myelin-derived axon-growth inhibition in the CNS.
Collapse
Affiliation(s)
- Jan M Schwab
- CNRS 7102, Equipe Développement Neuronal, Université Pierre et Marie Curie, 75005 Paris, France.
| | | | | |
Collapse
|
23
|
Abstract
STUDY DESIGN : Literature review. OBJECTIVE : The purpose of this paper is to review clinical treatment strategies and future developments in the treatment of acute spinal cord injury. SUMMARY OF BACKGROUND DATA : The treatment of acute spinal cord injury continues to be supportive. The search for specialized pharmacologic agents to prevent secondary injury and promote repair or regeneration remains heated. METHODS : Medline search from 1996 to present limited to clinical research and basic science review articles in the English Language. RESULTS : Steroids continue to be administered in the clinical setting of acute spinal cord injury primarily out of peer pressure and fear of litigation. Basic science experiments suggest that modulation of post-traumatic inflammation may provide the best opportunity to arrest the secondary injury cascade. Protein kinase and metalloproteinase inhibition are promising treatment strategies. Regeneration techniques are concentrating on cell transplantation and manipulating glial receptors and protein production. Clinical investigations are limited to Phase III trials on a very select few of these drugs. CONCLUSIONS : While many advances in the basic science of spinal cord injury provide optimism for future treatments, clinical science lags. At present, there are no pharmacologic strategies of proven benefit. Although steroids continue to be given to patients with spinal cord injury in many institutions, evidence of deleterious effects continues to accumulate. Current standard of care management includes support of arterial oxygenation and spinal cord perfusion pressure.
Collapse
Affiliation(s)
- R John Hurlbert
- From the University of Calgary Spine Program, Foothills Hospital and Medical Centre, Calgary, Alberta, Canada
| |
Collapse
|
24
|
Schwab JM, Brechtel K, Mueller CA, Failli V, Kaps HP, Tuli SK, Schluesener HJ. Experimental strategies to promote spinal cord regeneration--an integrative perspective. Prog Neurobiol 2006; 78:91-116. [PMID: 16487649 DOI: 10.1016/j.pneurobio.2005.12.004] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2005] [Revised: 12/13/2005] [Accepted: 12/13/2005] [Indexed: 11/17/2022]
Abstract
Detailed pathophysiological findings of secondary damage phenomena after spinal cord injury (SCI) as well as the identification of inhibitory and neurotrophic proteins have yielded a plethora of experimental therapeutic approaches. Main targets are (i) to minimize secondary damage progression (neuroprotection), (ii) to foster axon conduction (neurorestoration) and (iii) to supply a permissive environment to promote axonal sprouting (neuroregenerative therapies). Pre-clinical studies have raised hope in functional recovery through the antagonism of growth inhibitors, application of growth factors, cell transplantation, and vaccination strategies. To date, even though based on successful pre-clinical animal studies, results of clinical trials are characterized by dampened effects attributable to difficulties in the study design (patient heterogeneity) and species differences. A combination of complementary therapeutic strategies might be considered pre-requisite for future synergistic approaches. Here, we line out pre-clinical interventions resulting in improved functional neurological outcome after spinal cord injury and track them on their intended way to bedside.
Collapse
Affiliation(s)
- Jan M Schwab
- Institute of Brain Research, Calwer Str. 3, University of Tuebingen, Medical School, Calwerstr. 3, 72076 Tuebingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
25
|
Conrad S, Schluesener HJ, Trautmann K, Joannin N, Meyermann R, Schwab JM. Prolonged lesional expression of RhoA and RhoB following spinal cord injury. J Comp Neurol 2005; 487:166-75. [PMID: 15880494 DOI: 10.1002/cne.20561] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Inhibition of the small GTPase ras homology protein (Rho) or its downstream target, the Rho-associated kinase (ROCK), has been shown to promote axon regeneration and to improve functional recovery following spinal cord injury (SCI) in the adult rat. Here, we have analyzed the expression of RhoA and RhoB following spinal cord injury in order to assess whether Rho is a possible target for late pharmacological intervention. In control spinal cords, RhoA(+) cells were almost absent, whereas RhoB was localized to some ependymal cells, a few microglia, and some dissociated neurons. In injured spinal cords, RhoA(+) and RhoB(+)cells accumulated at perilesional areas and in the developing necrotic core early after injury at day 1. After reaching their maximum levels (RhoA at day 3; RhoB at day 1), RhoA(+) and RhoB(+) cell numbers remained significantly elevated until day 28. In areas remote from the lesion (> or =0.75 mm), a more discrete accumulation of RhoA(+) and RhoB(+) cells was observed, primarily in areas of ongoing Wallerian degeneration. RhoA and RhoB were predominantly expressed by polymorphonuclear granulocytes, ED1(+) microglia/macrophages, oligodendrocytes, some neurons, and swollen axons/neurites. Furthermore, expression was located to lesional, reactive astrocytes and fibroblastoid cells confined to areas of scar formation. Our experiments have determined that most RhoA(+) and RhoB(+) cells (>70%) are of mononuclear origin. The persistent presence of lesional RhoA(+) and RhoB(+) axon/neurite fibers over a period of 4 weeks after injury suggests that Rho inhibition is a putative therapeutic concept also for delayed intervention after SCI.
Collapse
Affiliation(s)
- Sabine Conrad
- Institute of Brain Research, D-72076 Tuebingen, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Ahmed Z, Dent RG, Leadbeater WE, Smith C, Berry M, Logan A. Matrix metalloproteases: degradation of the inhibitory environment of the transected optic nerve and the scar by regenerating axons. Mol Cell Neurosci 2005; 28:64-78. [PMID: 15607942 DOI: 10.1016/j.mcn.2004.08.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2004] [Accepted: 08/23/2004] [Indexed: 10/26/2022] Open
Abstract
After injury to the central nervous system, a glial/collagen scar forms at the lesion site, which is thought to act as a physicochemical barrier to regenerating axons. We have shown that scar formation in the transected optic nerve (ON) is attenuated when robust growth of axons is stimulated. Matrix metalloproteases (MMP), modulated by tissue inhibitors of MMP (TIMP), degrade a wide variety of extracellular matrix components (ECM) and may be activated by growing axons to remodel the ECM to allow regeneration through the inhibitory environment of the glial or collagen scar. Here, we investigate whether MMP levels are modulated in a nonregenerating (scarring) versus a regenerating (nonscarring) model of ON injury in vivo. Western blotting and immunohistochemistry revealed that MMP-1, -2, and -9 levels were higher and TIMP-1 and TIMP-2 levels were lower in regenerating compared to nonregenerating ON and retinae. In situ zymography demonstrated significantly greater MMP-related gelatinase activity in the regenerating model, mainly colocalized to astrocytes in the proximal ON stump and around the lesion site. These results suggest that activation of MMP and coincident down-regulation of TIMP may act to attenuate the inhibitory scarring in the regenerating ON, thus transforming the ON into a noninhibitory pathway for axon regrowth.
Collapse
Affiliation(s)
- Zubair Ahmed
- Molecular Neuroscience Group, Department of Medicine, University of Birmingham, Birmingham B15 2TT, UK.
| | | | | | | | | | | |
Collapse
|
27
|
Al Halabiah H, Delezoide AL, Cardona A, Moalic JM, Simonneau M. Expression pattern of NOGO and NgR genes during human development. Gene Expr Patterns 2005; 5:561-8. [PMID: 15749087 DOI: 10.1016/j.modgep.2004.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2004] [Revised: 10/25/2004] [Accepted: 10/26/2004] [Indexed: 12/16/2022]
Abstract
Nogo protein has been identified as the component of central nervous system (CNS) myelin that limits axonal regeneration. We investigated the expression of the genes encoding Nogo and its receptor, NgR, between weeks eight and 23 of human embryonic development, by quantitative radioactive in situ hybridization. At 8 weeks, we detected NOGO and NgR transcripts in developing neuronal and non-neuronal structures. We focused on two different structures: the brain and the dental germs. During this period of development, NOGO and NgR transcripts colocalized in the cortical and ventricular zones of the brain, with expression strongest for these two genes in the postmitotic cells of the cortical plate. In developing dental germs, NgR was more strongly expressed than NOGO at 16 and 21 weeks. NOGO and NgR were expressed in zones of epithelium-mesenchyme interaction, which induce the differentiation of ameloblasts/odontoblasts. These genes were expressed most strongly in differentiated cells.
Collapse
Affiliation(s)
- Hassan Al Halabiah
- Neurogénétique, INSERM E9935, Hôpital Robert Debré, AP-HP, 48 Boulevard Sérurier, 75019, Paris, France
| | | | | | | | | |
Collapse
|
28
|
Irving EA, Vinson M, Rosin C, Roberts JC, Chapman DM, Facci L, Virley DJ, Skaper SD, Burbidge SA, Walsh FS, Hunter AJ, Parsons AA. Identification of neuroprotective properties of anti-MAG antibody: a novel approach for the treatment of stroke? J Cereb Blood Flow Metab 2005; 25:98-107. [PMID: 15678116 DOI: 10.1038/sj.jcbfm.9600011] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The inhibitory activity of myelin-associated glycoprotein (MAG) on neurons is thought to contribute to the lack of regenerative capacity of the CNS after injury. The interaction of MAG and its neuronal receptors mediates bidirectional signaling between neurons and oligodendrocytes. The novel finding that an anti-MAG monoclonal antibody not only possesses the ability to neutralise the inhibitory effect of MAG on neurons but also directly protects oligodendrocytes from glutamate-mediated oxidative stress-induced cell death is reported here. Furthermore, administration of anti-MAG antibody (centrally and systemically) starting 1 hour after middle cerebral artery occlusion in the rat significantly reduced lesion volume at 7 days. This neuroprotection was associated with a robust improvement in motor function compared with animals receiving control IgG1. Together, these data highlight the potential for the use of anti-MAG antibodies as therapeutic agents for the treatment of stroke.
Collapse
Affiliation(s)
- Elaine A Irving
- Neurology & GI CEDD, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Harlow, Essex, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
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
|
30
|
Schnaar RL. Glycolipid-mediated cell-cell recognition in inflammation and nerve regeneration. Arch Biochem Biophys 2004; 426:163-72. [PMID: 15158667 DOI: 10.1016/j.abb.2004.02.019] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2003] [Revised: 02/12/2004] [Indexed: 12/19/2022]
Abstract
Cell surface complex carbohydrates have emerged as key recognition molecules, mediating physiological interactions between cells. Typically, glycans on one cell surface are engaged by complementary carbohydrate binding proteins (lectins) on an apposing cell, initiating appropriate cellular responses. Although many cell surface lectins have been identified in vertebrates, only a few of their endogenous carbohydrate ligands have been established. Each major class of cell surface glycans-glycoproteins, glycolipids, and proteoglycans-has been implicated as physiologically relevant lectin ligands. The current minireview focuses on findings that implicate glycosphingolipids as especially important molecules in cell-cell recognition in two different systems: the recognition of human leukocytes by E-selectin on the vascular endothelium during inflammation and the recognition of nerve cell axons by myelin-associated glycoprotein in myelin-axon stabilization and the regulation of axon regeneration.
Collapse
Affiliation(s)
- Ronald L Schnaar
- Departments of Pharmacology and Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
31
|
Brabeck C, Beschorner R, Conrad S, Mittelbronn M, Bekure K, Meyermann R, Schluesener HJ, Schwab JM. Lesional Expression of RhoA and RhoB following Traumatic Brain Injury in Humans. J Neurotrauma 2004; 21:697-706. [PMID: 15253798 DOI: 10.1089/0897715041269597] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inhibition of the small GTPase Rho or of its downstream target Rho-associated kinase (ROCK) has been shown to promote axon regeneration and to improve functional recovery following traumatic CNS lesions in the adult rat. In order to determine the expression pattern of RhoA and RhoB following human traumatic brain injury (TBI) and to assess whether Rho is a possible target for pharmacological intervention in humans, we investigated expression patterns of RhoA and RhoB in brain specimens from 25 patients who died after closed TBI in comparison to brain tissue derived from four neuropathologically unaffected control patients by immunohistochemistry. A highly significant lesional upregulation of both RhoA and RhoB was observed beginning several hours after the traumatic event and continuing for months after TBI. The cellular sources of both molecules included polymorphonuclear granulocytes, monocytes/macrophages, and reactive astrocytes. Additionally, expression of RhoA was also detected in neuronal cells in some of the cases. From our data, we conclude that inhibition of Rho is a promising mechanism for the development of new pharmacological interventions in human TBI. As the observed upregulation of RhoA and RhoB was still detectable months after TBI, we speculate that even delayed treatment with Rho inhibitors might be a therapeutic option.
Collapse
Affiliation(s)
- Christine Brabeck
- Institute of Brain Research, University of Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Song XY, Zhong JH, Wang X, Zhou XF. Suppression of p75NTR does not promote regeneration of injured spinal cord in mice. J Neurosci 2004; 24:542-6. [PMID: 14724254 PMCID: PMC6730005 DOI: 10.1523/jneurosci.4281-03.2004] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The neurotrophin receptor p75NTR is the coreceptor for Nogo receptor, mediating growth cone collapse in vitro by MAG, myelin oligodendrocyte glycoprotein (Omgp), and Nogo. Whether p75NTR plays any role in the failure of nerve regeneration in vivo is not known. Immunohistochemical data showed that p75NTR was expressed in only a very small subset of ascending sensory axons but not in any corticospinal axons in the dorsal column of either normal or injured spinal cord. Using p75NTR-deficient mice, we showed that the depletion of the functional p75NTR did not promote the regeneration of the descending corticospinal tract and ascending sensory neurons in the spinal cord 2 weeks after spinal cord injury. Local administration of p75NTR-Fc fusion molecule, the dominant-negative receptor to block the function of neurite outgrowth inhibitors, did not improve regeneration of ascending sensory neurons in the injured spinal cord. Our results suggest that p75NTR may not be a critical molecule mediating the function of myelin-associated inhibitory factors in vivo.
Collapse
Affiliation(s)
- Xing-Yun Song
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide 5001, Australia
| | | | | | | |
Collapse
|
33
|
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
|
34
|
Gentry JJ, Barker PA, Carter BD. The p75 neurotrophin receptor: multiple interactors and numerous functions. PROGRESS IN BRAIN RESEARCH 2004; 146:25-39. [PMID: 14699954 DOI: 10.1016/s0079-6123(03)46002-0] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The neurotrophin receptor p75 (p75NTR), is involved in a diverse array of cellular responses, including apoptosis, neurite outgrowth and myelination. Stimulation of p75NTR with neurotrophin can activate multiple downstream signals, including the small GTP binding protein Rac, the transcription factor NF-kappa B and the stress activated kinase, JNK. How these signals are generated and regulated to produce a specific cellular effect has yet to be fully elucidated. A number of proteins have recently been shown to interact with the intracellular domain of p75NTR. Here, we review these p75NTR interacting factors and the current evidence as to how they contribute to the functional effects of p75NTR activation.
Collapse
Affiliation(s)
- Jannifer J Gentry
- Center for Molecular Neuroscience, Department of Biochemistry, Vanderbilt University Medical School, Nashville, TN 37232, USA
| | | | | |
Collapse
|
35
|
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
|
36
|
Dubreuil CI, Winton MJ, McKerracher L. Rho activation patterns after spinal cord injury and the role of activated Rho in apoptosis in the central nervous system. J Cell Biol 2003; 162:233-43. [PMID: 12860969 PMCID: PMC2172802 DOI: 10.1083/jcb.200301080] [Citation(s) in RCA: 307] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Growth inhibitory proteins in the central nervous system (CNS) block axon growth and regeneration by signaling to Rho, an intracellular GTPase. It is not known how CNS trauma affects the expression and activation of RhoA. Here we detect GTP-bound RhoA in spinal cord homogenates and report that spinal cord injury (SCI) in both rats and mice activates RhoA over 10-fold in the absence of changes in RhoA expression. In situ Rho-GTP detection revealed that both neurons and glial cells showed Rho activation at SCI lesion sites. Application of a Rho antagonist (C3-05) reversed Rho activation and reduced the number of TUNEL-labeled cells by approximately 50% in both injured mouse and rat, showing a role for activated Rho in cell death after CNS injury. Next, we examined the role of the p75 neurotrophin receptor (p75NTR) in Rho signaling. After SCI, an up-regulation of p75NTR was detected by Western blot and observed in both neurons and glia. Treatment with C3-05 blocked the increase in p75NTR expression. Experiments with p75NTR-null mutant mice showed that immediate Rho activation after SCI is p75NTR dependent. Our results indicate that blocking overactivation of Rho after SCI protects cells from p75NTR-dependent apoptosis.
Collapse
Affiliation(s)
- Catherine I Dubreuil
- Département de pathologie et biologie cellulaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | | |
Collapse
|
37
|
Abstract
Developing neurons must respond to a wide range of extracellular signals during the process of brain morphogenesis. One mechanism through which immature neurons respond to such signals is by altering cellular actin dynamics. A recently discovered link between extracellular signaling events and the actin cytoskeleton is the WASP/WAVE (Wiscott-Aldrich Syndrome protein/WASP-family verprolin-homologous protein) family of proteins. Through a direct interaction with the Arp2/3 (actin-related protein) complex, this family functions to regulate the actin cytoskeleton by mediating signals from cdc42 as well as other small GTPases. To evaluate the role of WASP/WAVE proteins in the process of neuronal morphogenesis, we used a retroviral gene trap to generate a line of mice bearing a disruption in the WAVE1 gene. Using a heterologous reporter gene, we found that WAVE1 expression becomes increasingly restricted to the CNS over the course of development. Homozygous disruption of the WAVE1 gene results in postnatal lethality. In addition, these animals have severe limb weakness, a resting tremor, and notable neuroanatomical malformations without overt histopathology of peripheral organs. We did not detect any alterations in neuronal morphology in vivo or the ability of embryonic neurons to form processes in vitro. Our data indicate that WAVE1, although important for the general development of the CNS, is not essential for the formation and extension of neuritic processes.
Collapse
|
38
|
Kaplan DR, Miller FD. Axon growth inhibition: signals from the p75 neurotrophin receptor. Nat Neurosci 2003; 6:435-6. [PMID: 12715005 DOI: 10.1038/nn0503-435] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
39
|
Abstract
Nogo-A, a reticulon protein expressed by oligodendrocytes, contributes to the axonal growth inhibitory action of central myelin in growth cone collapse and neurite outgrowth in vitro assays, and antibody and inhibitor studies have implicated a role for Nogo in regeneration in the adult CNS in vivo. Three independent labs have now produced Nogo knockout mice with, quite unexpectedly, three different regeneration phenotypes.
Collapse
Affiliation(s)
- Clifford J Woolf
- Neural Plasticity Research Group, Department of Anesthesia and Critical Care, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
40
|
Josephson A, Trifunovski A, Schéele C, Widenfalk J, Wahlestedt C, Brené S, Olson L, Spenger C. Activity-induced and developmental downregulation of the Nogo receptor. Cell Tissue Res 2003; 311:333-42. [PMID: 12658441 DOI: 10.1007/s00441-002-0695-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2002] [Accepted: 12/11/2002] [Indexed: 11/27/2022]
Abstract
The three axon growth inhibitory proteins, myelin associated glycoprotein, oligodendrocyte-myelin glycoprotein and Nogo-A, can all bind to the Nogo-66 receptor (NgR). This receptor is expressed by neurons with high amounts in regions of high plasticity where Nogo expression is also high. We hypothesized that simultaneous presence of high levels of Nogo and its receptor in neurons confers a locked state to hippocampal and cortical microcircuitry and that one or both of these proteins must be effectively and temporarily downregulated to permit plastic structural changes underlying formation of long-term memory. Hence, we subjected rats to kainic acid treatment and exposed rats to running wheels and measured NgR mRNA levels by quantitative in situ hybridization at different time points. We also studied spinal cord injuries and quantified NgR mRNA levels in spinal cord and ganglia during a critical postnatal period using real-time PCR. Strikingly, kainic acid led to a strong transient downregulation of NgR mRNA levels in gyrus dentatus, hippocampus, and neocortex during a time when BDNF mRNA was upregulated instead. Animals exposed to running wheels for 3 and 7, but not 1 or 21, days showed a significant downregulation of NgR mRNA in cortex, hippocampus and the dentate gyrus. NgR mRNA levels decreased from high to low expression in spinal cord and ganglia during the first week of life. No robust regulation of NgR was observed in the spinal cord following spinal cord injury. Together, our data show that NgR levels in developing and adult neurons are regulated in vivo under different conditions. Strong, rapid and transient downregulation of NgR mRNA in response to kainic acid and after wheel running in cortex and hippocampus suggests a role for NgR and Nogo-A in plasticity, learning and memory.
Collapse
Affiliation(s)
- Anna Josephson
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Affiliation(s)
- Michael A Moskowitz
- Neuroscience Center, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Mass 02129, USA.
| | | |
Collapse
|
42
|
Abstract
Developing axons are guided to their targets by chemoattractive and chemorepulsive ligands. Ledda et al., in this issue of Neuron, demonstrate that the target-derived receptor glial cell line-derived neurotrophic factor receptor alpha1 (GFRalpha1) can also act in trans as an axon guidance molecule for neurons.
Collapse
|