1
|
Gallo G. The Axonal Actin Filament Cytoskeleton: Structure, Function, and Relevance to Injury and Degeneration. Mol Neurobiol 2024; 61:5646-5664. [PMID: 38216856 DOI: 10.1007/s12035-023-03879-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/13/2023] [Indexed: 01/14/2024]
Abstract
Early investigations of the neuronal actin filament cytoskeleton gave rise to the notion that, although growth cones exhibit high levels of actin filaments, the axon shaft exhibits low levels of actin filaments. With the development of new tools and imaging techniques, the axonal actin filament cytoskeleton has undergone a renaissance and is now an active field of research. This article reviews the current state of knowledge about the actin cytoskeleton of the axon shaft. The best understood forms of actin filament organization along axons are axonal actin patches and a submembranous system of rings that endow the axon with protrusive competency and structural integrity, respectively. Additional forms of actin filament organization along the axon have also been described and their roles are being elucidated. Extracellular signals regulate the axonal actin filament cytoskeleton and our understanding of the signaling mechanisms involved is being elaborated. Finally, recent years have seen advances in our perspective on how the axonal actin cytoskeleton is impacted by, and contributes to, axon injury and degeneration. The work to date has opened new venues and future research will undoubtedly continue to provide a richer understanding of the axonal actin filament cytoskeleton.
Collapse
Affiliation(s)
- Gianluca Gallo
- Department of Neural Sciences, Shriners Pediatric Research Center, Lewis Katz School of Medicine at Temple University, 3500 North Broad St, Philadelphia, PA, 19140, USA.
| |
Collapse
|
2
|
Lilienberg J, Hegyi Z, Szabó E, Hathy E, Málnási-Csizmadia A, Réthelyi JM, Apáti Á, Homolya L. Pharmacological Modulation of Neurite Outgrowth in Human Neural Progenitor Cells by Inhibiting Non-muscle Myosin II. Front Cell Dev Biol 2021; 9:719636. [PMID: 34604221 PMCID: PMC8484915 DOI: 10.3389/fcell.2021.719636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/27/2021] [Indexed: 12/31/2022] Open
Abstract
Studies on neural development and neuronal regeneration after injury are mainly based on animal models. The establishment of pluripotent stem cell (PSC) technology, however, opened new perspectives for better understanding these processes in human models by providing unlimited cell source for hard-to-obtain human tissues. Here, we aimed at identifying the molecular factors that confine and modulate an early step of neural regeneration, the formation of neurites in human neural progenitor cells (NPCs). Enhanced green fluorescent protein (eGFP) was stably expressed in NPCs differentiated from human embryonic and induced PSC lines, and the neurite outgrowth was investigated under normal and injury-related conditions using a high-content screening system. We found that inhibitors of the non-muscle myosin II (NMII), blebbistatin and its novel, non-toxic derivatives, initiated extensive neurite outgrowth in human NPCs. The extracellular matrix components strongly influenced the rate of neurite formation but NMII inhibitors were able to override the inhibitory effect of a restrictive environment. Non-additive stimulatory effect on neurite generation was also detected by the inhibition of Rho-associated, coiled-coil-containing protein kinase 1 (ROCK1), the upstream regulator of NMII. In contrast, inhibition of c-Jun N-terminal kinases (JNKs) had only a negligible effect, suggesting that the ROCK1 signal is dominantly manifested by actomyosin activity. In addition to providing a reliable cell-based in vitro model for identifying intrinsic mechanisms and environmental factors responsible for impeded axonal regeneration in humans, our results demonstrate that NMII and ROCK1 are important pharmacological targets for the augmentation of neural regeneration at the progenitor level. These studies may open novel perspectives for development of more effective pharmacological treatments and cell therapies for various neurodegenerative disorders.
Collapse
Affiliation(s)
- Julianna Lilienberg
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Zoltán Hegyi
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Eszter Szabó
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Edit Hathy
- Molecular Psychiatry and in vitro Disease Modelling Research Group, National Brain Research Project, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - András Málnási-Csizmadia
- MTA-ELTE Motor Pharmacology Research Group, Eötvös Loránd University, Budapest, Hungary.,Motorpharma, Ltd., Budapest, Hungary
| | - János M Réthelyi
- Molecular Psychiatry and in vitro Disease Modelling Research Group, National Brain Research Project, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Ágota Apáti
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - László Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
3
|
Porro C, Pennella A, Panaro MA, Trotta T. Functional Role of Non-Muscle Myosin II in Microglia: An Updated Review. Int J Mol Sci 2021; 22:ijms22136687. [PMID: 34206505 PMCID: PMC8267657 DOI: 10.3390/ijms22136687] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023] Open
Abstract
Myosins are a remarkable superfamily of actin-based motor proteins that use the energy derived from ATP hydrolysis to translocate actin filaments and to produce force. Myosins are abundant in different types of tissues and involved in a large variety of cellular functions. Several classes of the myosin superfamily are expressed in the nervous system; among them, non-muscle myosin II (NM II) is expressed in both neurons and non-neuronal brain cells, such as astrocytes, oligodendrocytes, endothelial cells, and microglia. In the nervous system, NM II modulates a variety of functions, such as vesicle transport, phagocytosis, cell migration, cell adhesion and morphology, secretion, transcription, and cytokinesis, as well as playing key roles during brain development, inflammation, repair, and myelination functions. In this review, we will provide a brief overview of recent emerging roles of NM II in resting and activated microglia cells, the principal regulators of immune processes in the central nervous system (CNS) in both physiological and pathological conditions. When stimulated, microglial cells react and produce a number of mediators, such as pro-inflammatory cytokines, free radicals, and nitric oxide, that enhance inflammation and contribute to neurodegenerative diseases. Inhibition of NM II could be a new therapeutic target to treat or to prevent CNS diseases.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Antonio Pennella
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
- Correspondence:
| |
Collapse
|
4
|
Liaci C, Camera M, Caslini G, Rando S, Contino S, Romano V, Merlo GR. Neuronal Cytoskeleton in Intellectual Disability: From Systems Biology and Modeling to Therapeutic Opportunities. Int J Mol Sci 2021; 22:ijms22116167. [PMID: 34200511 PMCID: PMC8201358 DOI: 10.3390/ijms22116167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/25/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Intellectual disability (ID) is a pathological condition characterized by limited intellectual functioning and adaptive behaviors. It affects 1–3% of the worldwide population, and no pharmacological therapies are currently available. More than 1000 genes have been found mutated in ID patients pointing out that, despite the common phenotype, the genetic bases are highly heterogeneous and apparently unrelated. Bibliomic analysis reveals that ID genes converge onto a few biological modules, including cytoskeleton dynamics, whose regulation depends on Rho GTPases transduction. Genetic variants exert their effects at different levels in a hierarchical arrangement, starting from the molecular level and moving toward higher levels of organization, i.e., cell compartment and functions, circuits, cognition, and behavior. Thus, cytoskeleton alterations that have an impact on cell processes such as neuronal migration, neuritogenesis, and synaptic plasticity rebound on the overall establishment of an effective network and consequently on the cognitive phenotype. Systems biology (SB) approaches are more focused on the overall interconnected network rather than on individual genes, thus encouraging the design of therapies that aim to correct common dysregulated biological processes. This review summarizes current knowledge about cytoskeleton control in neurons and its relevance for the ID pathogenesis, exploiting in silico modeling and translating the implications of those findings into biomedical research.
Collapse
Affiliation(s)
- Carla Liaci
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Mattia Camera
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Giovanni Caslini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Simona Rando
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Salvatore Contino
- Department of Engineering, University of Palermo, Viale delle Scienze Ed. 8, 90128 Palermo, Italy;
| | - Valentino Romano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze Ed. 16, 90128 Palermo, Italy;
| | - Giorgio R. Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
- Correspondence: ; Tel.: +39-0116706449; Fax: +39-0116706432
| |
Collapse
|
5
|
Hong ES, Yao HH, Min YJ, Sun J, Zhou X, Zeng XB, Yu W. The mechanism of electroacupuncture for treating spinal cord injury rats by mediating Rho/Rho-associated kinase signaling pathway. J Spinal Cord Med 2021; 44:364-374. [PMID: 31596180 PMCID: PMC8081320 DOI: 10.1080/10790268.2019.1665612] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Objective: To determine the changes of gene and protein expression through Rho/ROCK signaling pathway in EA treated spinal cord injury (SCI) rats and to unveil the possible underlying mechanism.Design: Animal study.Setting: Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine.Participants: Eighty Male Sprague Dawley rats.Interventions: Electroacupuncture at Yaoyangguan (GV3), Dazhui (GV14), Zusanli (ST36) and Ciliao (BL32) and/or blocking agent Y27632 treatment.Outcome Measures: Protein expression was detected by ELISA and Western blotting, mRNA expression was detected by quantitative PCR and in situ hybridization. Morphological changes in spinal cord were evaluated by HE-staining and Nissl staining. Hindlimb motor function in the rats was evaluated by Basso-Beattie-Bresnahan (BBB) assessment methods.Results: Compared with injured rats in SCI group, EA, blocking agent Y27632 and EA + blocking agent Y27632 treatment had significantly reduced mRNA and protein expression levels of RhoA and ROCKII, decreased p-MLC protein expression and p-MLC/MLC ratio, suppressed cPLA2 activity and PGE2 level, improved spinal cord tissue morphology and BBB score of lower limb movement function at 7 days and at 14 days (P < 0.01 or <0.05).Conclusion: Similar to the blocking agent Y27632, EA may have a notable inhibitory effect on the Rho/ROCK signaling pathway after SCI, therefore reducing the inhibition of axonal growth and inflammatory reaction may be a key mechanism of EA treatment for SCI.
Collapse
Affiliation(s)
- En-si Hong
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Hai-hua Yao
- Shanghai Eighth People’s Hospital, Shanghai, People’s Republic of China
| | - You-jiang Min
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China,Shanghai Eighth People’s Hospital, Shanghai, People’s Republic of China,Correspondence to: You-jiang Min, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, China and Shanghai Eighth People’s Hospital, Shanghai, 200235, People’s Republic of China
| | - Jie Sun
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Xuan Zhou
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Xue-bo Zeng
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Wan Yu
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| |
Collapse
|
6
|
Gyimesi M, Rauscher AÁ, Suthar SK, Hamow KÁ, Oravecz K, Lőrincz I, Borhegyi Z, Déri MT, Kiss ÁF, Monostory K, Szabó PT, Nag S, Tomasic I, Krans J, Tierney PJ, Kovács M, Kornya L, Málnási-Csizmadia A. Improved Inhibitory and Absorption, Distribution, Metabolism, Excretion, and Toxicology (ADMET) Properties of Blebbistatin Derivatives Indicate That Blebbistatin Scaffold Is Ideal for drug Development Targeting Myosin-2. J Pharmacol Exp Ther 2021; 376:358-373. [PMID: 33468641 DOI: 10.1124/jpet.120.000167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/07/2020] [Indexed: 11/22/2022] Open
Abstract
Blebbistatin, para-nitroblebbistatin (NBleb), and para-aminoblebbistatin (AmBleb) are highly useful tool compounds as they selectively inhibit the ATPase activity of myosin-2 family proteins. Despite the medical importance of the myosin-2 family as drug targets, chemical optimization has not yet provided a promising lead for drug development because previous structure-activity-relationship studies were limited to a single myosin-2 isoform. Here we evaluated the potential of blebbistatin scaffold for drug development and found that D-ring substitutions can fine-tune isoform specificity, absorption-distribution-metabolism-excretion, and toxicological properties. We defined the inhibitory properties of NBleb and AmBleb on seven different myosin-2 isoforms, which revealed an unexpected potential for isoform specific inhibition. We also found that NBleb metabolizes six times slower than blebbistatin and AmBleb in rats, whereas AmBleb metabolizes two times slower than blebbistatin and NBleb in human, and that AmBleb accumulates in muscle tissues. Moreover, mutagenicity was also greatly reduced in case of AmBleb. These results demonstrate that small substitutions have beneficial functional and pharmacological consequences, which highlight the potential of the blebbistatin scaffold for drug development targeting myosin-2 family proteins and delineate a route for defining the chemical properties of further derivatives to be developed. SIGNIFICANCE STATEMENT: Small substitutions on the blebbistatin scaffold have beneficial functional and pharmacological consequences, highlighting their potential in drug development targeting myosin-2 family proteins.
Collapse
Affiliation(s)
- Máté Gyimesi
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Anna Á Rauscher
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Sharad Kumar Suthar
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Kamirán Á Hamow
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Kinga Oravecz
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - István Lőrincz
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Zsolt Borhegyi
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Máté T Déri
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Ádám F Kiss
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Katalin Monostory
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Pál Tamás Szabó
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Suman Nag
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Ivan Tomasic
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Jacob Krans
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Patrick J Tierney
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Mihály Kovács
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - László Kornya
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - András Málnási-Csizmadia
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| |
Collapse
|
7
|
Costa AR, Sousa MM. Non-Muscle Myosin II in Axonal Cell Biology: From the Growth Cone to the Axon Initial Segment. Cells 2020; 9:cells9091961. [PMID: 32858875 PMCID: PMC7563147 DOI: 10.3390/cells9091961] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
By binding to actin filaments, non-muscle myosin II (NMII) generates actomyosin networks that hold unique contractile properties. Their dynamic nature is essential for neuronal biology including the establishment of polarity, growth cone formation and motility, axon growth during development (and axon regeneration in the adult), radial and longitudinal axonal tension, and synapse formation and function. In this review, we discuss the current knowledge on the spatial distribution and function of the actomyosin cytoskeleton in different axonal compartments. We highlight some of the apparent contradictions and open questions in the field, including the role of NMII in the regulation of axon growth and regeneration, the possibility that NMII structural arrangement along the axon shaft may control both radial and longitudinal contractility, and the mechanism and functional purpose underlying NMII enrichment in the axon initial segment. With the advances in live cell imaging and super resolution microscopy, it is expected that in the near future the spatial distribution of NMII in the axon, and the mechanisms by which it participates in axonal biology will be further untangled.
Collapse
|
8
|
Conventional and Non-Conventional Roles of Non-Muscle Myosin II-Actin in Neuronal Development and Degeneration. Cells 2020; 9:cells9091926. [PMID: 32825197 PMCID: PMC7566000 DOI: 10.3390/cells9091926] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Myosins are motor proteins that use chemical energy to produce mechanical forces driving actin cytoskeletal dynamics. In the brain, the conventional non-muscle myosin II (NMII) regulates actin filament cytoskeletal assembly and contractile forces during structural remodeling of axons and dendrites, contributing to morphology, polarization, and migration of neurons during brain development. NMII isoforms also participate in neurotransmission and synaptic plasticity by driving actin cytoskeletal dynamics during synaptic vesicle release and retrieval, and formation, maturation, and remodeling of dendritic spines. NMIIs are expressed differentially in cerebral non-neuronal cells, such as microglia, astrocytes, and endothelial cells, wherein they play key functions in inflammation, myelination, and repair. Besides major efforts to understand the physiological functions and regulatory mechanisms of NMIIs in the nervous system, their contributions to brain pathologies are still largely unclear. Nonetheless, genetic mutations or deregulation of NMII and its regulatory effectors are linked to autism, schizophrenia, intellectual disability, and neurodegeneration, indicating non-conventional roles of NMIIs in cellular mechanisms underlying neurodevelopmental and neurodegenerative disorders. Here, we summarize the emerging biological roles of NMIIs in the brain, and discuss how actomyosin signaling contributes to dysfunction of neurons and glial cells in the context of neurological disorders. This knowledge is relevant for a deep understanding of NMIIs on the pathogenesis and therapeutics of neuropsychiatric and neurodegenerative diseases.
Collapse
|
9
|
Wang G, Wang T, Hu Y, Wang J, Wang Y, Zhang Y, Li F, Liu W, Sun Y, Yu B, Kou J. NMMHC IIA triggers neuronal autophagic cell death by promoting F-actin-dependent ATG9A trafficking in cerebral ischemia/reperfusion. Cell Death Dis 2020; 11:428. [PMID: 32513915 PMCID: PMC7280511 DOI: 10.1038/s41419-020-2639-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022]
Abstract
Previous findings have shown that non-muscle myosin heavy-chain IIA (NMMHC IIA) is involved in autophagy induction triggered by starvation in D. melanogaster; however, its functional contribution to neuronal autophagy remains unclear. The aim of this study is to explore the function of NMMHC IIA in cerebral ischemia-induced neuronal autophagy and the underlying mechanism related to autophagy-related gene 9A (ATG9A) trafficking. Functional assays and molecular mechanism studies were used to investigate the role of NMMHC IIA in cerebral ischemia-induced neuronal autophagy in vivo and in vitro. A middle cerebral artery occlusion (MCAO) model in mice was used to evaluate the therapeutic effect of blebbistatin, a myosin II ATPase inhibitor. Herein, either depletion or knockdown of NMMHC IIA led to increased cell viability in both primary cultured cortical neurons and pheochromocytoma (PC12) cells exposed to oxygen–glucose deprivation/reoxygenation (OGD/R). In addition, NMMHC IIA and autophagic marker LC3B were upregulated by OGD/R, and inhibition of NMMHC IIA significantly reduced OGD-induced neuronal autophagy. Furthermore, NMMHC IIA-induced autophagy is through its interactions with F-actin and ATG9A in response to OGD/R. The NMMHC IIA–actin interaction contributes to ATG9A trafficking and autophagosome formation. Inhibition of the NMMHC IIA–actin interaction using blebbistatin and the F-actin polymerization inhibitor cytochalasin D significantly suppressed ATG9A trafficking and autophagy induction. Furthermore, blebbistatin significantly improved neurological deficits and infarct volume after ischemic attack in mice, accompanied by ATG9A trafficking and autophagy inhibition. These findings demonstrate neuroprotective effects of NMMHC IIA inhibition on regulating ATG9A trafficking-dependent autophagy activation in the context of cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Guangyun Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Tiezheng Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yang Hu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jieman Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yan Wang
- Department of Neurology, University of California, Davis, School of Medicine and Shriners Hospital, Sacramento, CA, 95817, Berkeley, USA
| | - Yuanyuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Fang Li
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Wentao Liu
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 210029, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Deparment of Biotechnology and Pharmaceutical Sciences, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Boyang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Resource and Developmemt of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Junping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
10
|
Neurite regrowth stimulation by a red-light spot focused on the neuronal cell soma following blue light-induced retraction. Sci Rep 2019; 9:18210. [PMID: 31796850 PMCID: PMC6890775 DOI: 10.1038/s41598-019-54687-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 11/18/2019] [Indexed: 11/13/2022] Open
Abstract
The interaction of light with biological tissues has been considered for various therapeutic applications. Light-induced neurite growth has the potential to be a clinically useful technique for neuron repair. However, most previous studies used either a large illumination area to accelerate overall neurite growth or employed a light spot to guide a growing neurite. It is not clear if optical stimulation can induce the regrowth of a retracted neurite. In the present work, we used blue light (wavelength: 473 nm) to cause neurite retraction, and we proved that using a red-light (wavelength: 650 nm) spot to illuminate the soma near the junction of the retracted neurite could induce neurite regrowth. As a comparison, we found that green light (wavelength 550 nm) had a 62% probability of inducing neurite regrowth, while red light had a 75% probability of inducing neurite regrowth at the same power level. Furthermore, the neurite regrowth length induced by red light was increased by the pre-treatment with inhibitors of myosin functions. We also observed actin propagation from the soma to the tip of the re-growing neurite following red-light stimulation of the soma. The red light-induced extension and regrowth were abrogated in the calcium-free medium. These results suggest that illumination with a red-light spot on the soma may trigger the regrowth of a neurite after the retraction caused by blue-light illumination.
Collapse
|
11
|
Javier-Torrent M, Marco S, Rocandio D, Pons-Vizcarra M, Janes PW, Lackmann M, Egea J, Saura CA. Presenilin/γ-secretase-dependent EphA3 processing mediates axon elongation through non-muscle myosin IIA. eLife 2019; 8:43646. [PMID: 31577226 PMCID: PMC6774734 DOI: 10.7554/elife.43646] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 09/18/2019] [Indexed: 01/08/2023] Open
Abstract
EphA/ephrin signaling regulates axon growth and guidance of neurons, but whether this process occurs also independently of ephrins is unclear. We show that presenilin-1 (PS1)/γ-secretase is required for axon growth in the developing mouse brain. PS1/γ-secretase mediates axon growth by inhibiting RhoA signaling and cleaving EphA3 independently of ligand to generate an intracellular domain (ICD) fragment that reverses axon defects in PS1/γ-secretase- and EphA3-deficient hippocampal neurons. Proteomic analysis revealed that EphA3 ICD binds to non-muscle myosin IIA (NMIIA) and increases its phosphorylation (Ser1943), which promotes NMIIA filament disassembly and cytoskeleton rearrangement. PS1/γ-secretase-deficient neurons show decreased phosphorylated NMIIA and NMIIA/actin colocalization. Moreover, pharmacological NMII inhibition reverses axon retraction in PS-deficient neurons suggesting that NMIIA mediates PS/EphA3-dependent axon elongation. In conclusion, PS/γ-secretase-dependent EphA3 cleavage mediates axon growth by regulating filament assembly through RhoA signaling and NMIIA, suggesting opposite roles of EphA3 on inhibiting (ligand-dependent) and promoting (receptor processing) axon growth in developing neurons.
Collapse
Affiliation(s)
- Míriam Javier-Torrent
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sergi Marco
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Daniel Rocandio
- Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Lleida, Spain
| | - Maria Pons-Vizcarra
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Peter W Janes
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Martin Lackmann
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Joaquim Egea
- Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Lleida, Spain
| | - Carlos A Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Xiao WP, Ding LLQ, Min YJ, Yang HY, Yao HH, Sun J, Zhou X, Zeng XB, Yu W. Electroacupuncture Promoting Axonal Regeneration in Spinal Cord Injury Rats via Suppression of Nogo/NgR and Rho/ROCK Signaling Pathway. Neuropsychiatr Dis Treat 2019; 15:3429-3442. [PMID: 31997879 PMCID: PMC6918258 DOI: 10.2147/ndt.s216874] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To observe the changes of Nogo/NgR and Rho/ROCK signaling pathway-related gene and protein expression in rats with spinal cord injury (SCI) treated with electroacupuncture (EA) and to further investigate the possible mechanism of EA for treating SCI. METHODS Allen's method was used to create the SCI rat model. Sixty-four model rats were further subdivided into four subgroups, namely, the SCI model group (SCI), EA treatment group (EA), blocking agent Y27632 treatment group (Y27632) and EA+blocking agent Y27632 treatment group (EA+Y), according to the treatment received. The rats were subjected to EA and/or blocking agent Y27632 treatment. After 14 days, injured spinal cord tissue was extracted for analysis. The mRNA and protein expression levels were determined by real-time fluorescence quantitative PCR and Western blotting, respectively. Cell apoptosis changes in the spinal cord were evaluated by in situ hybridization. Hindlimb motor function in the rats was evaluated by Basso-Beattie-Bresnahan assessment methods. RESULTS Except for RhoA protein expression, compared with the SCI model group, EA, blocking agent Y27632 and EA+blocking agent Y27632 treatment groups had significantly reduced mRNA and protein expression of Nogo-A, NgR, LINGO-1, RhoA and ROCK II in spinal cord tissues, increased mRNA and protein expression of MLCP, decreased p-MYPT1 protein expression and p-MYPT1/MYPT1 ratio, and caspase3 expression, and improved lower limb movement function after treatment for 14 days (P<0.01 or <0.05). The combination of EA and the blocking agent Y27632 was superior to EA or blocking agent Y27632 treatment alone (P < 0.01 or <0.05). CONCLUSION EA may have an obvious inhibitory effect on the Nogo/NgR and Rho/ROCK signaling pathway after SCI, thereby reducing the inhibition of axonal growth, which may be a key mechanism of EA treatment for SCI.
Collapse
Affiliation(s)
- Wei-Ping Xiao
- Spinal Department of Orthopedics and Department of Acupuncture, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Li-Li-Qiang Ding
- Department of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - You-Jiang Min
- Spinal Department of Orthopedics and Department of Acupuncture, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, People's Republic of China.,Department of Traditional Chinese Medicine, Shanghai Eighth People's Hospital, Shanghai, People's Republic of China
| | - Hua-Yuan Yang
- Institute of Traditional Chinese Medicine Engineering, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Hai-Hua Yao
- Department of Traditional Chinese Medicine, Shanghai Eighth People's Hospital, Shanghai, People's Republic of China
| | - Jie Sun
- Spinal Department of Orthopedics and Department of Acupuncture, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Xuan Zhou
- Spinal Department of Orthopedics and Department of Acupuncture, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Xue-Bo Zeng
- Spinal Department of Orthopedics and Department of Acupuncture, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Wan Yu
- Spinal Department of Orthopedics and Department of Acupuncture, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, People's Republic of China
| |
Collapse
|
13
|
Miller KE, Suter DM. An Integrated Cytoskeletal Model of Neurite Outgrowth. Front Cell Neurosci 2018; 12:447. [PMID: 30534055 PMCID: PMC6275320 DOI: 10.3389/fncel.2018.00447] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/07/2018] [Indexed: 12/27/2022] Open
Abstract
Neurite outgrowth underlies the wiring of the nervous system during development and regeneration. Despite a significant body of research, the underlying cytoskeletal mechanics of growth and guidance are not fully understood, and the relative contributions of individual cytoskeletal processes to neurite growth are controversial. Here, we review the structural organization and biophysical properties of neurons to make a semi-quantitative comparison of the relative contributions of different processes to neurite growth. From this, we develop the idea that neurons are active fluids, which generate strong contractile forces in the growth cone and weaker contractile forces along the axon. As a result of subcellular gradients in forces and material properties, actin flows rapidly rearward in the growth cone periphery, and microtubules flow forward in bulk along the axon. With this framework, an integrated model of neurite outgrowth is proposed that hopefully will guide new approaches to stimulate neuronal growth.
Collapse
Affiliation(s)
- Kyle E Miller
- Department of Integrative Biology, Michigan State University, East Lansing, MI, United States
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.,Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States.,Birck Nanotechnology Center, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
14
|
Li M, Wen Y, Zhang R, Xie F, Zhang G, Qin X. Adenoviral vector-induced silencing of RGMa attenuates blood-brain barrier dysfunction in a rat model of MCAO/reperfusion. Brain Res Bull 2018; 142:54-62. [PMID: 29935233 DOI: 10.1016/j.brainresbull.2018.06.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 06/15/2018] [Accepted: 06/18/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Repulsive guidance molecule A (RGMa) is implicated in focal cerebral ischemia-reperfusion (I/R) injury, but its mechanisms are still largely unknown. This work focused on the effects of RGMa on the blood-brain barrier (BBB) after focal cerebral I/R injury. METHODS Sprague-Dawley (SD) rats were randomly divided into four groups: sham, middle cerebral artery occlusion (MCAO)/reperfusion (I/R), MCAO/reperfusion administered recombinant adenovirus expressing sh-con (I/R + sh-con) and MCAO/reperfusion administered recombinant adenovirus expressing sh-RGMa (I/R + sh-RGMa) groups. Infarct volume, brain edema and neurological scores were evaluated at 3 day after reperfusion. Evens blue leakage and transmission electron microscopy was performed. And the expression level of claudin-5 and ZO-1, CDC-42 and PAK-1, RGMa were detected by western blot. RESULTS Compared with I/R or I/R + sh-con groups, I/R + sh-RGMa group showed smaller infarction volume, attenuated brain edema, improved neurological scores and better BBB integrity, such as reduced Evans blue leakage and ultra-structural change. We also observed improved BBB function followed by down-regulation of MMP-9 and up-regulation of claudin-5 and ZO-1 in the I/R + sh-RGMa group. In addition, up-regulation of the CDC-42 and PAK-1 in the I/R + sh-RGMa group was obtained. CONCLUSIONS RGMa may be involved in I/R injury associated with BBB dysfunction via the CDC-42/PAK-1 signal pathway and may be a promising therapeutic target for I/R injury.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, Inner Mongolia People's Hospital, Hohhot, China
| | - Yuetao Wen
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Rongrong Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fei Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
15
|
Sherman SP, Bang AG. High-throughput screen for compounds that modulate neurite growth of human induced pluripotent stem cell-derived neurons. Dis Model Mech 2018; 11:dmm.031906. [PMID: 29361516 PMCID: PMC5894944 DOI: 10.1242/dmm.031906] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/29/2017] [Indexed: 01/01/2023] Open
Abstract
Development of technology platforms to perform compound screens of human induced pluripotent stem cell (hiPSC)-derived neurons with relatively high throughput is essential to realize their potential for drug discovery. Here, we demonstrate the feasibility of high-throughput screening of hiPSC-derived neurons using a high-content, image-based approach focused on neurite growth, a process that is fundamental to formation of neural networks and nerve regeneration. From a collection of 4421 bioactive small molecules, we identified 108 hit compounds, including 37 approved drugs, that target molecules or pathways known to regulate neurite growth, as well as those not previously associated with this process. These data provide evidence that many pathways and targets known to play roles in neurite growth have similar activities in hiPSC-derived neurons that can be identified in an unbiased phenotypic screen. The data also suggest that hiPSC-derived neurons provide a useful system to study the mechanisms of action and off-target activities of the approved drugs identified as hits, leading to a better understanding of their clinical efficacy and toxicity, especially in the context of specific human genetic backgrounds. Finally, the hit set we report constitutes a sublibrary of approved drugs and tool compounds that modulate neurites. This sublibrary will be invaluable for phenotypic analyses and interrogation of hiPSC-based disease models as probes for defining phenotypic differences and cellular vulnerabilities in patient versus control cells, as well as for investigations of the molecular mechanisms underlying human neurite growth in development and maintenance of neuronal networks, and nerve regeneration. Summary: High-throughput, small molecule screening of hiPSC-derived neurons using a high-content, image-based approach focused on neurite growth identified hit compounds, including approved drugs, which target molecules or pathways known to regulate neurite growth.
Collapse
Affiliation(s)
- Sean P Sherman
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute La Jolla, CA 92037, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute La Jolla, CA 92037, USA
| |
Collapse
|
16
|
Repulsive Guidance Molecule a (RGMa) Induces Neuropathological and Behavioral Changes That Closely Resemble Parkinson's Disease. J Neurosci 2017; 37:9361-9379. [PMID: 28842419 DOI: 10.1523/jneurosci.0084-17.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 07/12/2017] [Accepted: 08/11/2017] [Indexed: 01/06/2023] Open
Abstract
Repulsive guidance molecule member a (RGMa) is a membrane-associated or released guidance molecule that is involved in axon guidance, cell patterning, and cell survival. In our previous work, we showed that RGMa is significantly upregulated in the substantia nigra of patients with Parkinson's disease. Here we demonstrate the expression of RGMa in midbrain human dopaminergic (DA) neurons. To investigate whether RGMa might model aspects of the neuropathology of Parkinson's disease in mouse, we targeted RGMa to adult midbrain dopaminergic neurons using adeno-associated viral vectors. Overexpression of RGMa resulted in a progressive movement disorder, including motor coordination and imbalance, which is typical for a loss of DA release in the striatum. In line with this, RGMa induced selective degeneration of dopaminergic neurons in the substantia nigra (SN) and affected the integrity of the nigrostriatal system. The degeneration of dopaminergic neurons was accompanied by a strong microglia and astrocyte activation. The behavioral, molecular, and anatomical changes induced by RGMa in mice are remarkably similar to the clinical and neuropathological hallmarks of Parkinson's disease. Our data indicate that dysregulation of RGMa plays an important role in the pathology of Parkinson's disease, and antibody-mediated functional interference with RGMa may be a disease modifying treatment option.SIGNIFICANCE STATEMENT Parkinson's disease (PD) is a neurodegenerative disease characterized by severe motor dysfunction due to progressive degeneration of mesencephalic dopaminergic (DA) neurons in the substantia nigra. To date, there is no regenerative treatment available. We previously showed that repulsive guidance molecule member a (RGMa) is upregulated in the substantia nigra of PD patients. Adeno-associated virus-mediated targeting of RGMa to mouse DA neurons showed that overexpression of this repulsive axon guidance and cell patterning cue models the behavioral and neuropathological characteristics of PD in a remarkable way. These findings have implications for therapy development as interfering with the function of this specific axon guidance cue may be beneficial to the survival of DA neurons.
Collapse
|
17
|
Tao T, Feng JZ, Xu GH, Fu J, Li XG, Qin XY. Minocycline Promotes Neurite Outgrowth of PC12 Cells Exposed to Oxygen-Glucose Deprivation and Reoxygenation Through Regulation of MLCP/MLC Signaling Pathways. Cell Mol Neurobiol 2017; 37:417-426. [PMID: 27098315 DOI: 10.1007/s10571-016-0374-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/17/2015] [Indexed: 10/21/2022]
Abstract
Minocycline, a semi-synthetic second-generation derivative of tetracycline, has been reported to exert neuroprotective effects both in animal models and in clinic trials of neurological diseases. In the present study, we first investigated the protective effects of minocycline on oxygen-glucose deprivation and reoxygenation-induced impairment of neurite outgrowth and its potential mechanism in the neuronal cell line, PC12 cells. We found that minocycline significantly increased cell viability, promoted neurite outgrowth and enhanced the expression of growth-associated protein-43 (GAP-43) in PC12 cells exposed to oxygen-glucose deprivation/reoxygenation injury. In addition, immunoblots revealed that minocycline reversed the overexpression of phosphorylated myosin light chain (MLC) and the suppression of activated extracellular signal-regulated kinase 1/2 (ERK1/2) caused by oxygen-glucose deprivation/reoxygenation injury. Moreover, the minocycline-induced neurite outgrowth was significantly blocked by Calyculin A (1 nM), an inhibitor of myosin light chain phosphatase (MLCP), but not by an ERK1/2 inhibitor (U0126; 10 μM). These findings suggested that minocycline activated the MLCP/MLC signaling pathway in PC12 cells after oxygen-glucose deprivation/reoxygenation injury, which resulted in the promotion of neurite outgrowth.
Collapse
Affiliation(s)
- Tao Tao
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, 64600, Sichuan Province, China
| | - Jin-Zhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guang-Hui Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jie Fu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, 64600, Sichuan Province, China
| | - Xiao-Gang Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, 64600, Sichuan Province, China
| | - Xin-Yue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
18
|
Omotade OF, Pollitt SL, Zheng JQ. Actin-based growth cone motility and guidance. Mol Cell Neurosci 2017; 84:4-10. [PMID: 28268126 DOI: 10.1016/j.mcn.2017.03.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/27/2017] [Accepted: 03/03/2017] [Indexed: 11/27/2022] Open
Abstract
Nerve growth cones, the dilated tip of developing axons, are equipped with exquisite abilities to sense environmental cues and to move rapidly through complex terrains of developing brain, leading the axons to their specific targets for precise neuronal wiring. The actin cytoskeleton is the major component of the growth cone that powers its directional motility. Past research has provided significant insights into the mechanisms by which growth cones translate extracellular signals into directional migration. In this review, we summarize the actin-based mechanisms underlying directional growth cone motility, examine novel findings, and discuss the outstanding questions concerning the actin-based growth cone behaviors.
Collapse
Affiliation(s)
- Omotola F Omotade
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States; Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Stephanie L Pollitt
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States; Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - James Q Zheng
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States; Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322, United States; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, United States.
| |
Collapse
|
19
|
Tsai SYA, Su TP. Sigma-1 Receptors Fine-Tune the Neuronal Networks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:79-83. [PMID: 28315266 PMCID: PMC6100794 DOI: 10.1007/978-3-319-50174-1_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The endoplasmic reticular (ER) protein sigma-1 receptor (Sig-1R) has been implicated in CNS disorders including but not limited to neurodegenerative diseases, depression , amnesia, and substance abuse. Sig-1Rs are particularly enriched in the specific domain where ER membranes make contacts with the mitochondria (MAM). Within that specific domain, Sig-1Rs play significant roles governing calcium signaling and reactive oxygen species homeostasis to maintain proper neuronal functions. Studies showed that the Sig-1R is pivotal to regulate neuroplasticity and neural survival via multiple aspects of mechanism. Numerous reports have been focusing on Sig-1R's regulatory effects in ER stress, mitochondrial function, oxidative stress and protein chaperoning. In this book chapter, we will discuss the emerging role of Sig-1R in balancing the populations of neuron and glia and their implications in CNS diseases.
Collapse
Affiliation(s)
- Shang-Yi Anne Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, 21224, USA.
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, US Department of Health and Human Services, Baltimore, MD, 21224, USA
| |
Collapse
|
20
|
Bye N, Christie KJ, Turbic A, Basrai HS, Turnley AM. Rho kinase inhibition following traumatic brain injury in mice promotes functional improvement and acute neuron survival but has little effect on neurogenesis, glial responses or neuroinflammation. Exp Neurol 2016; 279:86-95. [DOI: 10.1016/j.expneurol.2016.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 12/27/2022]
|
21
|
Xu X, Gao Y, Zhai Z, Zhang S, Shan F, Feng J. Repulsive guidance molecule a blockade exerts the immunoregulatory function in DCs stimulated with ABP and LPS. Hum Vaccin Immunother 2016; 12:2169-2180. [PMID: 26986456 DOI: 10.1080/21645515.2016.1164361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Repulsive guidance molecule a (RGMa) is an axonal guidance molecule that has recently found to exert function in immune system. This study evaluated the function of RGMa in modulation of dendritic cells (DCs) function stimulated with Achyranthes bidentata polysaccharide (ABP) and lipopolysaccharide (LPS) using a RGMa-neutralizing antibody. Compared with the Control-IgG/ABP and Control-IgG/LPS groups, DCs in the Anti-RGMa/ABP and Anti-RGMa/LPS groups 1) showed small, round cells with a few cell processes and organelles, and many pinocytotic vesicles; 2) had decreased MHC II, CD86, CD80, and CD40 expression; 3) displayed the decreased IL-12p70, IL-1β and TNF-α levels and increased IL-10 secretion; 4) had a high percentage of FITC-dextran uptake; and 5) displayed a reduced ability to drive T cell proliferation and reinforced T cell polarization toward a Th2 cytokine pattern. We conclude that DCs treated with RGMa-neutralizing antibodies present with tolerogenic and immunoregulatory characteristics, which provides new insights into further understanding of the function of RGMa.
Collapse
Affiliation(s)
- Xuxu Xu
- a Department of Neurology, Shengjing Hospital , Affiliated Hospital of China Medical University , Shenyang , PR China
| | - Yan Gao
- a Department of Neurology, Shengjing Hospital , Affiliated Hospital of China Medical University , Shenyang , PR China
| | - Zhiyong Zhai
- a Department of Neurology, Shengjing Hospital , Affiliated Hospital of China Medical University , Shenyang , PR China
| | - Shuo Zhang
- a Department of Neurology, Shengjing Hospital , Affiliated Hospital of China Medical University , Shenyang , PR China
| | - Fengping Shan
- b Department of Immunology, School of Basic Medical Science , China Medical University , Shenyang , PR China
| | - Juan Feng
- a Department of Neurology, Shengjing Hospital , Affiliated Hospital of China Medical University , Shenyang , PR China
| |
Collapse
|
22
|
Abstract
A spinal cord injury refers to an injury to the spinal cord that is caused by a trauma instead of diseases. Spinal cord injury includes a primary mechanical injury and a much more complex secondary injury process involving inflammation, oxidation, excitotoxicity, and cell death. During the secondary injury, many signal pathways are activated and play important roles in mediating the pathogenesis of spinal cord injury. Among them, the RhoA/Rho kinase pathway plays a particular role in mediating spinal degeneration and regeneration. In this review, we will discuss the role and mechanism of RhoA/Rho kinase-mediated spinal cord pathogenesis, as well as the potential of targeting RhoA/Rho kinase as a strategy for promoting both neuroprotection and axonal regeneration.
Collapse
Affiliation(s)
- Xiangbing Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
23
|
Harada S, Matsuura W, Takano M, Tokuyama S. Withdrawal: Proteomic Profiling in the Spinal Cord and Sciatic Nerve in a Global Cerebral Ischemia-Induced Mechanical Allodynia Mouse Model. Biol Pharm Bull 2016; 39:230-8. [DOI: 10.1248/bpb.b15-00647] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shinichi Harada
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University
| | - Wataru Matsuura
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University
| | - Masaoki Takano
- Department of Life Sciences Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University
| | - Shogo Tokuyama
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University
| |
Collapse
|
24
|
Menon S, Gupton SL. Building Blocks of Functioning Brain: Cytoskeletal Dynamics in Neuronal Development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:183-245. [PMID: 26940519 PMCID: PMC4809367 DOI: 10.1016/bs.ircmb.2015.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural connectivity requires proper polarization of neurons, guidance to appropriate target locations, and establishment of synaptic connections. From when neurons are born to when they finally reach their synaptic partners, neurons undergo constant rearrangment of the cytoskeleton to achieve appropriate shape and polarity. Of particular importance to neuronal guidance to target locations is the growth cone at the tip of the axon. Growth-cone steering is also dictated by the underlying cytoskeleton. All these changes require spatiotemporal control of the cytoskeletal machinery. This review summarizes the proteins that are involved in modulating the actin and microtubule cytoskeleton during the various stages of neuronal development.
Collapse
Affiliation(s)
- Shalini Menon
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America; Neuroscience Center and Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC, United States of America; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America.
| |
Collapse
|
25
|
Newell-Litwa KA, Horwitz R, Lamers ML. Non-muscle myosin II in disease: mechanisms and therapeutic opportunities. Dis Model Mech 2015; 8:1495-515. [PMID: 26542704 PMCID: PMC4728321 DOI: 10.1242/dmm.022103] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The actin motor protein non-muscle myosin II (NMII) acts as a master regulator of cell morphology, with a role in several essential cellular processes, including cell migration and post-synaptic dendritic spine plasticity in neurons. NMII also generates forces that alter biochemical signaling, by driving changes in interactions between actin-associated proteins that can ultimately regulate gene transcription. In addition to its roles in normal cellular physiology, NMII has recently emerged as a critical regulator of diverse, genetically complex diseases, including neuronal disorders, cancers and vascular disease. In the context of these disorders, NMII regulatory pathways can be directly mutated or indirectly altered by disease-causing mutations. NMII regulatory pathway genes are also increasingly found in disease-associated copy-number variants, particularly in neuronal disorders such as autism and schizophrenia. Furthermore, manipulation of NMII-mediated contractility regulates stem cell pluripotency and differentiation, thus highlighting the key role of NMII-based pharmaceuticals in the clinical success of stem cell therapies. In this Review, we discuss the emerging role of NMII activity and its regulation by kinases and microRNAs in the pathogenesis and prognosis of a diverse range of diseases, including neuronal disorders, cancer and vascular disease. We also address promising clinical applications and limitations of NMII-based inhibitors in the treatment of these diseases and the development of stem-cell-based therapies.
Collapse
Affiliation(s)
- Karen A Newell-Litwa
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rick Horwitz
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Marcelo L Lamers
- Department of Morphological Sciences, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-010, Brazil
| |
Collapse
|
26
|
Sayyad WA, Amin L, Fabris P, Ercolini E, Torre V. The role of myosin-II in force generation of DRG filopodia and lamellipodia. Sci Rep 2015; 5:7842. [PMID: 25598228 PMCID: PMC4648386 DOI: 10.1038/srep07842] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 12/15/2014] [Indexed: 11/10/2022] Open
Abstract
Differentiating neurons process the mechanical stimulus by exerting the protrusive forces through lamellipodia and filopodia. We used optical tweezers, video imaging and immunocytochemistry to analyze the role of non-muscle myosin-II on the protrusive force exerted by lamellipodia and filopodia from developing growth cones (GCs) of isolated Dorsal Root Ganglia (DRG) neurons. When the activity of myosin-II was inhibited by 30 μM Blebbistatin protrusion/retraction cycles of lamellipodia slowed down and during retraction lamellipodia could not lift up axially as in control condition. Inhibition of actin polymerization with 25 nM Cytochalasin-D and of microtubule polymerization with 500 nM Nocodazole slowed down the protrusion/retraction cycles, but only Cytochalasin-D decreased lamellipodia axial motion. The force exerted by lamellipodia treated with Blebbistatin decreased by 50%, but, surprisingly, the force exerted by filopodia increased by 20-50%. The concomitant disruption of microtubules caused by Nocodazole abolished the increase of the force exerted by filopodia treated with Blebbistatin. These results suggest that; i- Myosin-II controls the force exerted by lamellipodia and filopodia; ii- contractions of the actomyosin complex formed by filaments of actin and myosin have an active role in ruffle formation; iii- myosin-II is an essential component of the structural stability of GCs architecture.
Collapse
Affiliation(s)
- Wasim A Sayyad
- Neuroscience Area, International School for Advanced Studies (SISSA), IT-34136 Trieste, Italy
| | - Ladan Amin
- Neuroscience Area, International School for Advanced Studies (SISSA), IT-34136 Trieste, Italy
| | - Paolo Fabris
- Neuroscience Area, International School for Advanced Studies (SISSA), IT-34136 Trieste, Italy
| | - Erika Ercolini
- Neuroscience Area, International School for Advanced Studies (SISSA), IT-34136 Trieste, Italy
| | - Vincent Torre
- Neuroscience Area, International School for Advanced Studies (SISSA), IT-34136 Trieste, Italy
| |
Collapse
|
27
|
Song MY, Tian FF, Wang YZ, Huang X, Guo JL, Ding DX. Potential roles of the RGMa-FAK-Ras pathway in hippocampal mossy fiber sprouting in the pentylenetetrazole kindling model. Mol Med Rep 2014; 11:1738-44. [PMID: 25420768 PMCID: PMC4270322 DOI: 10.3892/mmr.2014.2993] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 08/01/2014] [Indexed: 11/23/2022] Open
Abstract
Mossy fiber sprouting (MFS) is a unique feature of chronic epilepsy. However, the molecular signals underlying MFS are still unclear. The repulsive guidance molecule A (RGMa) appears to contribute to axon growth and axonal guidance, and may exert its biological effects by dephosphorylating focal adhesion kinase (FAK) at Tyr397, then regulating the activation of Ras. The objective of this study was to explore the expression patterns of RGMa, FAK (Tyr397) and Ras in epileptogenesis, and their correlation with MFS. The epileptic models were established by intraperitoneal pentylenetetrazole (PTZ) injection of Sprague-Dawley rats. At 3 days and at 1, 2, 4 and 6 weeks after the first PTZ injection, Timm staining was scored at different time points in the CA3 region of the hippocampus and dentate gyrus. The protein levels of RGMa, FAK (Tyr397) and Ras were analyzed at different time points in the CA3 region of the hippocampus using immunofluorescence, immunohistochemistry and western blot analysis. Compared with the control (saline-injected) group, the expression of RGMa in the CA3 area was significantly downregulated (P<0.05) from 3 days and still maintained the low expression at 6 weeks in the PTZ group. The expression of FAK (Tyr397) and Ras was upregulated (P<0.05) in the PTZ groups. The Timm score in the CA3 region was significantly higher than that in the control group at different time points and reached a peak at 4 weeks. In the CA3 region, no obvious distinction was observed at the different time points in the control group. To the best of our knowledge, these are the first results to indicate that the RGMa-FAK-Ras pathway may be involved in MFS and the development of temporal lobe epilepsy.
Collapse
Affiliation(s)
- Ming-Yu Song
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Fa-Fa Tian
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yu-Zhong Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xia Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jia-Ling Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Dong-Xue Ding
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
28
|
Fujita Y, Yamashita T. Axon growth inhibition by RhoA/ROCK in the central nervous system. Front Neurosci 2014; 8:338. [PMID: 25374504 PMCID: PMC4205828 DOI: 10.3389/fnins.2014.00338] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/06/2014] [Indexed: 12/31/2022] Open
Abstract
Rho kinase (ROCK) is a serine/threonine kinase and a downstream target of the small GTPase Rho. The RhoA/ROCK pathway is associated with various neuronal functions such as migration, dendrite development, and axonal extension. Evidence from animal studies reveals that RhoA/ROCK signaling is involved in various central nervous system (CNS) diseases, including optic nerve and spinal cord injuries, stroke, and neurodegenerative diseases. Given that RhoA/ROCK plays a critical role in the pathophysiology of CNS diseases, the development of therapeutic agents targeting this pathway is expected to contribute to the treatment of CNS diseases. The RhoA/ROCK pathway mediates the effects of myelin-associated axon growth inhibitors—Nogo, myelin-associated glycoprotein (MAG), oligodendrocyte-myelin glycoprotein (OMgp), and repulsive guidance molecule (RGM). Blocking RhoA/ROCK signaling can reverse the inhibitory effects of these molecules on axon outgrowth, and promotes axonal sprouting and functional recovery in animal models of CNS injury. To date, several RhoA/ROCK inhibitors have been under development or in clinical trials as therapeutic agents for neurological disorders. In this review, we focus on the RhoA/ROCK signaling pathway in neurological disorders. We also discuss the potential therapeutic approaches of RhoA/ROCK inhibitors for various neurological disorders.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University Osaka, Japan ; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology Tokyo, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University Osaka, Japan ; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology Tokyo, Japan
| |
Collapse
|
29
|
Soluble guanylate cyclase generation of cGMP regulates migration of MGE neurons. J Neurosci 2013; 33:16897-914. [PMID: 24155296 DOI: 10.1523/jneurosci.1871-13.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Here we have provided evidence that nitric oxide-cyclic GMP (NO-cGMP) signaling regulates neurite length and migration of immature neurons derived from the medial ganglionic eminence (MGE). Dlx1/2(-/-) and Lhx6(-/-) mouse mutants, which exhibit MGE interneuron migration defects, have reduced expression of the gene encoding the α subunit of a soluble guanylate cyclase (Gucy1A3). Furthermore, Dlx1/2(-/-) mouse mutants have reduced expression of NO synthase 1 (NOS1). Gucy1A3(-/-) mice have a transient reduction in cortical interneuron number. Pharmacological inhibition of soluble guanylate cyclase and NOS activity rapidly induces neurite retraction of MGE cells in vitro and in slice culture and robustly inhibits cell migration from the MGE and caudal ganglionic eminence. We provide evidence that these cellular phenotypes are mediated by activation of the Rho signaling pathway and inhibition of myosin light chain phosphatase activity.
Collapse
|
30
|
CRMP-2 is involved in axon growth inhibition induced by RGMa in vitro and in vivo. Mol Neurobiol 2012; 47:903-13. [PMID: 23275173 DOI: 10.1007/s12035-012-8385-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 12/04/2012] [Indexed: 10/27/2022]
Abstract
Repulsive guidance molecule-a (RGMa) is associated with axon growth inhibition in different central nervous system (CNS) injuries, but its signaling pathways remain unclear. We examined the involvement of collapsin response mediator protein-2 (CRMP-2), a common downstream target of Rho-kinase and GSK-3β, in vitro by culturing neonatal rat primary cortical neurons with RGMa protein, Rho-kinase inhibitor (Y-27632), and GSK-3β inhibitor. We examined CRMP-2 in vivo by suppressing RGMa expression using recombinant adenovirus (rAd-shRGMa) in a rat MCAO/reperfusion model. RGMa induced neurite retraction and CRMP-2 phosphorylation in vitro, which were reversed by either Rho-kinase or GSK-3β inhibitors. After MCAO/reperfusion in rats, pCRMP-2 protein was greatly increased in the ischemic cortex, axons were damaged severely, Neurofilament-200 (NF-200) expression was significantly decreased, and neurological deficits were significant, which were all improved by down-regulating RGMa. We concluded RGMa inhibits axon growth by phosphorylating CRMP-2 via both Rho-kinase and GSK-3β signaling pathways.
Collapse
|
31
|
Lah GJ, Key B. Dual roles of the chemorepellent axon guidance molecule RGMa in establishing pioneering axon tracts and neural fate decisions in embryonic vertebrate forebrain. Dev Neurobiol 2012; 72:1458-70. [DOI: 10.1002/dneu.22010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 12/01/2011] [Accepted: 01/19/2012] [Indexed: 11/11/2022]
|
32
|
Yu P, Santiago LY, Katagiri Y, Geller HM. Myosin II activity regulates neurite outgrowth and guidance in response to chondroitin sulfate proteoglycans. J Neurochem 2012; 120:1117-28. [PMID: 22191382 PMCID: PMC3296867 DOI: 10.1111/j.1471-4159.2011.07638.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are major components of the extracellular matrix in the CNS that inhibit axonal regeneration after CNS injury. Signaling pathways in neurons triggered by CSPGs are still largely unknown. In this study, using well-characterized in vitro assays for neurite outgrowth and neurite guidance, we demonstrate a major role for myosin II in the response of neurons to CSPGs. We found that the phosphorylation of myosin II regulatory light chains is increased by CSPGs. Specific inhibition of myosin II activity with blebbistatin allows growing neurites to cross onto CSPG-rich areas and increases the length of neurites of neurons growing on CSPGs. Using specific gene knockdown, we demonstrate selective roles for myosin IIA and IIB in these processes. Time lapse microscopy and immunocytochemistry demonstrated that CSPGs also inhibit cell adhesion and cell spreading. Inhibition of myosin II selectively accelerated neurite initiation without altering cell adhesion and spreading on CSPGs.
Collapse
Affiliation(s)
- Panpan Yu
- Developmental Neurobiology Section, Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
33
|
Kubo T, Tokita S, Yamashita T. Repulsive guidance molecule-a and demyelination: implications for multiple sclerosis. J Neuroimmune Pharmacol 2011; 7:524-8. [PMID: 22183806 DOI: 10.1007/s11481-011-9334-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 12/06/2011] [Indexed: 12/23/2022]
Abstract
Drug development for neurodegenerative and neuroinflammatory diseases such as multiple sclerosis and traumatic brain injury is challenging. One promising strategy is to target a molecule with multiple biological actions affecting divergent pathophysiological disease phases simultaneously since these diseases arise from multiple pathological phases. In recent years, we pursued this strategy with a focus on multiple sclerosis and spinal cord injury and found that repulsive guidance molecule-a (RGMa) inhibits regeneration of injured CNS axons following spinal cord injury. We also found that RGMa enhances CD4(+) T cell activation facilitating CNS demyelination in an animal model of MS, mouse experimental autoimmune encephalomyelitis (EAE), which supports the idea that RGMa has distinct pathological actions. The multiple functions of RGMa in the CNS and the immune system would provide a therapeutic opportunity to concurrently block the autoimmune reactions and axon injury in neurodegenerative and neuroinflammatory diseases. In this article, we introduce the therapeutic potential of targeting RGMa as a novel intervention for MS and spinal cord injury.
Collapse
Affiliation(s)
- Takekazu Kubo
- Molecular Function and Pharmacology Laboratories, Pharmaceutical Business, Taisho Pharmaceutical Co., Ltd, 403, Yoshino-cho 1-Chome, Kita-ku, Saitama-shi, Saitama, 331-9530, Japan.
| | | | | |
Collapse
|
34
|
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: 32] [Impact Index Per Article: 2.5] [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
|
35
|
Kubo T, Yamaguchi A, Iwata N, Yamashita T. The therapeutic effects of Rho-ROCK inhibitors on CNS disorders. Ther Clin Risk Manag 2011; 4:605-15. [PMID: 18827856 PMCID: PMC2500253 DOI: 10.2147/tcrm.s2907] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Rho-kinase (ROCK) is a serine/threonine kinase and one of the major downstream effectors of the small GTPase Rho. The Rho-ROCK pathway is involved in many aspects of neuronal functions including neurite outgrowth and retraction. The Rho-ROCK pathway becomes an attractive target for the development of drugs for treating central nervous system (CNS) disorders, since it has been recently revealed that this pathway is closely related to the pathogenesis of several CNS disorders such as spinal cord injuries, stroke, and Alzheimer's disease (AD). In the adult CNS, injured axons regenerate poorly due to the presence of myelin-associated axonal growth inhibitors such as myelin-associated glycoprotein (MAG), Nogo, oligodendrocyte-myelin glycoprotein (OMgp), and the recently identified repulsive guidance molecule (RGM). The effects of these inhibitors are reversed by blockade of the Rho-ROCK pathway in vitro, and the inhibition of this pathway promotes axonal regeneration and functional recovery in the injured CNS in vivo. In addition, the therapeutic effects of the Rho-ROCK inhibitors have been demonstrated in animal models of stroke. In this review, we summarize the involvement of the Rho-ROCK pathway in CNS disorders such as spinal cord injuries, stroke, and AD and also discuss the potential of Rho-ROCK inhibitors in the treatment of human CNS disorders.
Collapse
Affiliation(s)
- Takekazu Kubo
- Department of Neurobiology, Graduate School of Medicine, Chiba University 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | | | | | | |
Collapse
|
36
|
Expression, phosphorylation, and glycosylation of CNS proteins in aversive operant conditioning associated memory in Lymnaea stagnalis. Neuroscience 2011; 186:94-109. [DOI: 10.1016/j.neuroscience.2011.04.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 04/08/2011] [Accepted: 04/09/2011] [Indexed: 11/18/2022]
|
37
|
Suter DM, Miller KE. The emerging role of forces in axonal elongation. Prog Neurobiol 2011; 94:91-101. [PMID: 21527310 DOI: 10.1016/j.pneurobio.2011.04.002] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 03/18/2011] [Accepted: 04/06/2011] [Indexed: 11/26/2022]
Abstract
An understanding of how axons elongate is needed to develop rational strategies to treat neurological diseases and nerve injury. Growth cone-mediated neuronal elongation is currently viewed as occurring through cytoskeletal dynamics involving the polymerization of actin and tubulin subunits at the tip of the axon. However, recent work suggests that axons and growth cones also generate forces (through cytoskeletal dynamics, kinesin, dynein, and myosin), forces induce axonal elongation, and axons lengthen by stretching. This review highlights results from various model systems (Drosophila, Aplysia, Xenopus, chicken, mouse, rat, and PC12 cells), supporting a role for forces, bulk microtubule movements, and intercalated mass addition in the process of axonal elongation. We think that a satisfying answer to the question, "How do axons grow?" will come by integrating the best aspects of biophysics, genetics, and cell biology.
Collapse
Affiliation(s)
- Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907-2054, United States.
| | | |
Collapse
|
38
|
Dent EW, Gupton SL, Gertler FB. The growth cone cytoskeleton in axon outgrowth and guidance. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a001800. [PMID: 21106647 DOI: 10.1101/cshperspect.a001800] [Citation(s) in RCA: 402] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Axon outgrowth and guidance to the proper target requires the coordination of filamentous (F)-actin and microtubules (MTs), the dynamic cytoskeletal polymers that promote shape change and locomotion. Over the past two decades, our knowledge of the many guidance cues, receptors, and downstream signaling cascades involved in neuronal outgrowth and guidance has increased dramatically. Less is known, however, about how those cascades of information converge and direct appropriate remodeling and interaction of cytoskeletal polymers, the ultimate effectors of movement and guidance. During development, much of the communication that occurs between environmental guidance cues and the cytoskeleton takes place at the growing tip of the axon, the neuronal growth cone. Several articles on this topic focus on the "input" to the growth cone, the myriad of receptor types, and their corresponding cognate ligands. Others investigate the signaling cascades initiated by receptors and propagated by second messenger pathways (i.e., kinases, phosphatases, GTPases). Ultimately, this plethora of information converges on proteins that associate directly with the actin and microtubule cytoskeletons. The role of these cytoskeletal-associated proteins, as well as the cytoskeleton itself in axon outgrowth and guidance, is the subject of this article.
Collapse
Affiliation(s)
- Erik W Dent
- Department of Anatomy, University of Wisconsin-Madison, 53706, USA
| | | | | |
Collapse
|
39
|
Bridgman PC. Myosin motor proteins in the cell biology of axons and other neuronal compartments. Results Probl Cell Differ 2010; 48:91-105. [PMID: 19554282 DOI: 10.1007/400_2009_10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Most neurons of both the central and peripheral nervous systems express multiple members of the myosin superfamily that include nonmuscle myosin II, and a number of classes of unconventional myosins. Several classes of unconventional myosins found in neurons have been shown to play important roles in transport processes. A general picture of the myosin-dependent transport processes in neurons is beginning to emerge, although much more work still needs to be done to fully define these roles and establish the importance of myosin for axonal transport. Myosins appear to contribute to three types of transport processes in neurons; recycling of receptors or other membrane components, dynamic tethering of vesicular components, and transport or tethering of protein translational machinery including mRNA. Defects in one or more of these functions have potential to contribute to disease processes.
Collapse
Affiliation(s)
- Paul C Bridgman
- Department of Anatomy and Neurobiology, Box 8108, Washington University School of Medicine, 660 Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
40
|
Nohra R, Beyeen AD, Guo JP, Khademi M, Sundqvist E, Hedreul MT, Sellebjerg F, Smestad C, Oturai AB, Harbo HF, Wallström E, Hillert J, Alfredsson L, Kockum I, Jagodic M, Lorentzen J, Olsson T. RGMA and IL21R show association with experimental inflammation and multiple sclerosis. Genes Immun 2010; 11:279-93. [PMID: 20072140 DOI: 10.1038/gene.2009.111] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Rat chromosome 1 harbors overlapping quantitative trait loci (QTL) for cytokine production and experimental models of inflammatory diseases. We fine-dissected this region that regulated cytokine production, myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE), anti-MOG antibodies and pristane-induced arthritis (PIA) in advanced intercross lines (AILs). Analysis in the tenth and twelfth generation of AILs resolved the region in two narrow QTL, Eae30 and Eae31. Eae30 showed linkage to MOG-EAE, anti-MOG antibodies and levels of interleukin-6 (IL-6). Eae31 showed linkage to EAE, PIA, anti-MOG antibodies and levels of tumor necrosis factor (TNF) and IL-6. Confidence intervals defined a limited set of potential candidate genes, with the most interesting being RGMA, IL21R and IL4R. We tested the association with multiple sclerosis (MS) in a Nordic case-control material. A single nucleotide polymorphism in RGMA associated with MS in males (odds ratio (OR)=1.33). Polymorphisms of RGMA also correlated with changes in the expression of interferon-gamma (IFN-gamma) and TNF in cerebrospinal fluid of MS patients. In IL21R, there was one positively associated (OR=1.14) and two protective (OR=0.87 and 0.68) haplotypes. One of the protective haplotypes correlated to lower IFN-gamma expression in peripheral blood mononuclear cells of MS patients. We conclude that RGMA and IL21R and their pathways are crucial in MS pathogenesis and warrant further studies as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- R Nohra
- Department of Clinical Neuroscience, Neuroimmunology Unit, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Jiang XS, Wassif CA, Backlund PS, Song L, Holtzclaw LA, Li Z, Yergey AL, Porter FD. Activation of Rho GTPases in Smith-Lemli-Opitz syndrome: pathophysiological and clinical implications. Hum Mol Genet 2010; 19:1347-57. [PMID: 20067919 DOI: 10.1093/hmg/ddq011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Smith-Lemli-Opitz syndrome (SLOS) is a malformation syndrome with neurocognitive deficits due to mutations of DHCR7 that impair the reduction of 7-dehydrocholesterol to cholesterol. To investigate the pathological processes underlying the neurocognitive deficits, we compared protein expression in Dhcr7(+/+) and Dhcr7(Delta3-5/Delta3-5) brain tissue. One of the proteins identified was cofilin-1, an actin depolymerizing factor which regulates neuronal dendrite and axon formation. Differential expression of cofilin-1 was due to increased phosphorylation. Phosphorylation of cofilin-1 is regulated by Rho GTPases through Rho-Rock-Limk-Cofilin-1 and Rac/Cdc42-Pak-Limk-Cofilin-1 pathways. Pull-down assays were used to demonstrate increased activation of RhoA, Rac1 and Cdc42 in Dhcr7(Delta3-5/Delta3-5) brains. Consistent with increased activation of these Rho GTPases, we observed increased phosphorylation of both Limk and Pak in mutant brain tissue. Altered Rho/Rac signaling impairs normal dendritic and axonal formation, and mutations in genes encoding regulators and effectors of the Rho GTPases underlie other human mental retardation syndromes. Thus, we hypothesized that aberrant activation of Rho/Rac could have functional consequences for dendrite and axonal growth. In vitro analysis of Dhcr7(Delta3-5/Delta3-5) hippocampal neurons demonstrated both axonal and dendritic abnormalities. Developmental abnormalities of neuronal process formation may contribute to the neurocognitive deficits found in SLOS and may represent a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Xiao-Sheng Jiang
- Section on Molecular Dysmorphology, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Peng X, Zhou Z, Hu J, Fink DJ, Mata M. Soluble Nogo receptor down-regulates expression of neuronal Nogo-A to enhance axonal regeneration. J Biol Chem 2009; 285:2783-95. [PMID: 19901030 DOI: 10.1074/jbc.m109.046425] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Nogo-A, a member of the reticulon family, is present in neurons and oligodendrocytes. Nogo-A in central nervous system (CNS) myelin prevents axonal regeneration through interaction with Nogo receptor 1, but the function of Nogo-A in neurons is less known. We found that after axonal injury, Nogo-A is increased in dorsal root ganglion (DRG) neurons unable to regenerate following a dorsal root injury or a sciatic nerve ligation-cut injury and that exposure in vitro to CNS myelin dramatically enhanced neuronal Nogo-A mRNA and protein through activation of RhoA while inhibiting neurite growth. Knocking down neuronal Nogo-A by small interfering RNA results in a marked increase of neurite outgrowth. We constructed a nonreplicating herpes simplex virus vector (QHNgSR) to express a truncated soluble fragment of Nogo receptor 1 (NgSR). NgSR released from QHNgSR prevented myelin inhibition of neurite extension by hippocampal and DRG neurons in vitro. NgSR prevents RhoA activation by myelin and decreases neuronal Nogo-A. Subcutaneous inoculation of QHNgSR to transduce DRG neurons resulted in improved regeneration of myelinated fibers in both the dorsal root and the spinal dorsal root entry zone, with concomitant improvement in sensory behavior. The results indicate that neuronal Nogo-A is an important intermediate in neurite growth dynamics and its expression is regulated by signals related to axonal injury and regeneration, that CNS myelin appears to activate signaling events that mimic axonal injury, and that NgSR released from QHNgSR may be used to improve recovery after injury.
Collapse
Affiliation(s)
- Xiangmin Peng
- Department of Neurology, University of Michigan, and Ann Arbor Veterans Affairs Healthcare System, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
43
|
Inutsuka A, Goda M, Fujiyoshi Y. Calyculin A-induced neurite retraction is critically dependent on actomyosin activation but not on polymerization state of microtubules. Biochem Biophys Res Commun 2009; 390:1160-6. [PMID: 19857460 DOI: 10.1016/j.bbrc.2009.10.108] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Accepted: 10/21/2009] [Indexed: 11/30/2022]
Abstract
Calyculin A (CL-A), a toxin isolated from the marine sponge Discodermia calyx, is a strong inhibitor of protein phosphatase 1 (PP1) and 2A (PP2A). Although CL-A is known to induce rapid neurite retraction in developing neurons, the cytoskeletal dynamics of this retraction have remained unclear. Here, we investigated the cytoskeletal dynamics during CL-A-induced neurite retraction in cultured rat hippocampal neurons, using fluorescence microscopy as well as polarized light microscopy, which can visualize the polymerization state of the cytoskeleton in living cells. We observed that MTs were bent while maintaining their polymerization state during the neurite retraction. In addition, we also found that CL-A still induced neurite retraction when MTs were depolymerized by nocodazole or stabilized by paclitaxel. These results imply a mechanism other than depolymerization of MTs for CL-A-induced neurite retraction. Our pharmacological studies showed that blebbistatin and cytochalasin D, an inhibitor of myosin II and a depolymerizer of actin, strongly inhibited CL-A-induced neurite retraction. Based on all these findings, we propose that CL-A generates strong contractile forces by actomyosin to induce rapid neurite retraction independently from MT depolymerization.
Collapse
Affiliation(s)
- Ayumu Inutsuka
- Department of Biophysics, Graduate School of Science, Kyoto University, Oiwake, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | |
Collapse
|
44
|
Nash M, Pribiag H, Fournier AE, Jacobson C. Central nervous system regeneration inhibitors and their intracellular substrates. Mol Neurobiol 2009; 40:224-35. [PMID: 19763907 DOI: 10.1007/s12035-009-8083-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 08/27/2009] [Indexed: 12/11/2022]
Abstract
Injury to the central nervous system (CNS) initiates a cascade of responses that is inhibitory to the regeneration of neurons and full recovery. At the site of injury, glial cells conspire with an inhibitory biochemical milieu to construct both physical and chemical barriers that prevent the outgrowth of axons to or beyond the lesion site. These inhibitors include factors derived from myelin, repulsive guidance cues, and chondroitin sulfate proteoglycans. Each bind receptors on the axon surface to initiating intracellular signaling cascades that ultimately result in cytoskeletal reorganization and growth cone collapse. Here, we present an overview of the molecules, receptors, and signaling pathways that inhibit CNS regeneration, with a particular focus on the intracellular signaling machinery that may function as convergent targets for multiple inhibitory ligands.
Collapse
Affiliation(s)
- Michelle Nash
- Department of Biology, University of Waterloo, ON, Canada
| | | | | | | |
Collapse
|
45
|
Norman LL, Stroka K, Aranda-Espinoza H. Guiding Axons in the Central Nervous System: A Tissue Engineering Approach. TISSUE ENGINEERING PART B-REVIEWS 2009; 15:291-305. [DOI: 10.1089/ten.teb.2009.0114] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Leann L. Norman
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Kimberly Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Helim Aranda-Espinoza
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
46
|
Ronen D, Ravid S. Myosin II tailpiece determines its paracrystal structure, filament assembly properties, and cellular localization. J Biol Chem 2009; 284:24948-57. [PMID: 19553683 DOI: 10.1074/jbc.m109.023754] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Non muscle myosin II (NMII) is a major motor protein present in all cell types. The three known vertebrate NMII isoforms share high sequence homology but play different cellular roles. The main difference in sequence resides in the C-terminal non-helical tailpiece (tailpiece). In this study we demonstrate that the tailpiece is crucial for proper filament size, overcoming the intrinsic properties of the coiled-coil rod. Furthermore, we show that the tailpiece by itself determines the NMII filament structure in an isoform-specific manner, thus providing a possible mechanism by which each NMII isoform carries out its unique cellular functions. We further show that the tailpiece determines the cellular localization of NMII-A and NMII-B and is important for NMII-C role in focal adhesion complexes. We mapped NMII-C sites phosphorylated by protein kinase C and casein kinase II and showed that these phosphorylations affect its solubility properties and cellular localization. Thus phosphorylation fine-tunes the tailpiece effects on the coiled-coil rod, enabling dynamic regulation of NMII-C assembly. We thus show that the small tailpiece of NMII is a distinct domain playing a role in isoform-specific filament assembly and cellular functions.
Collapse
Affiliation(s)
- Daniel Ronen
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research, Israel-Canada, the Hebrew University, Hadassah Medical School, Jerusalem 91220, Israel
| | | |
Collapse
|
47
|
Kollins KM, Hu J, Bridgman PC, Huang YQ, Gallo G. Myosin-II negatively regulates minor process extension and the temporal development of neuronal polarity. Dev Neurobiol 2009; 69:279-98. [PMID: 19224562 DOI: 10.1002/dneu.20704] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The earliest stage in the development of neuronal polarity is characterized by extension of undifferentiated "minor processes" (MPs), which subsequently differentiate into the axon and dendrites. We investigated the role of the myosin II motor protein in MP extension using forebrain and hippocampal neuron cultures. Chronic treatment of neurons with the myosin II ATPase inhibitor blebbistatin increased MP length, which was also seen in myosin IIB knockouts. Through live-cell imaging, we demonstrate that myosin II inhibition triggers rapid minor process extension to a maximum length range. Myosin II activity is determined by phosphorylation of its regulatory light chains (rMLC) and mediated by myosin light chain kinase (MLCK) or RhoA-kinase (ROCK). Pharmacological inhibition of MLCK or ROCK increased MP length moderately, with combined inhibition of these kinases resulting in an additive increase in MP length similar to the effect of direct inhibition of myosin II. Selective inhibition of RhoA signaling upstream of ROCK, with cell-permeable C3 transferase, increased both the length and number of MPs. To determine whether myosin II affected development of neuronal polarity, MP differentiation was examined in cultures treated with direct or indirect myosin II inhibitors. Significantly, inhibition of myosin II, MLCK, or ROCK accelerated the development of neuronal polarity. Increased myosin II activity, through constitutively active MLCK or RhoA, decreased both the length and number of MPs and, consequently, delayed or abolished the development of neuronal polarity. Together, these data indicate that myosin II negatively regulates MP extension, and the developmental time course for axonogenesis.
Collapse
Affiliation(s)
- K M Kollins
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, USA.
| | | | | | | | | |
Collapse
|
48
|
Inactivation of Ras by p120GAP via focal adhesion kinase dephosphorylation mediates RGMa-induced growth cone collapse. J Neurosci 2009; 29:6649-62. [PMID: 19458235 DOI: 10.1523/jneurosci.0927-09.2009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The repulsive guidance molecule RGMa performs several functions in the developing and adult CNSs. RGMa, through its receptor neogenin, induces growth cone collapse and neurite outgrowth inhibition. Here, we demonstrate that RGMa binding to neogenin leads to the inactivation of Ras, which is required for the RGMa-mediated repulsive function in cortical neurons. This signal transduction is mediated by the Ras-specific GTPase-activating protein (GAP) p120GAP. The SH2 domain of p120GAP interacts with focal adhesion kinase (FAK), which is phosphorylated at Tyr-397. When the cells are stimulated with RGMa, FAK undergoes dephosphorylation at Tyr-397 and is dissociated from p120GAP, and this dissociation is followed by an increase in the interaction between p120GAP and GTP-Ras. In addition, the knockdown of p120GAP prevents RGMa-induced growth cone collapse and neurite outgrowth inhibition. Furthermore, RGMa stimulation induces Akt inactivation through p120GAP, and the expression of the constitutively active Akt prevents RGMa-induced growth cone collapse. Thus, RGMa binding to neogenin regulates p120GAP activity through FAK Tyr-397 dephosphorylation, leading to the inactivation of Ras and its downstream effector Akt, and this signal transduction plays a role in the RGMa-mediated repulsive function.
Collapse
|
49
|
Hata K, Kaibuchi K, Inagaki S, Yamashita T. Unc5B associates with LARG to mediate the action of repulsive guidance molecule. ACTA ACUST UNITED AC 2009; 184:737-50. [PMID: 19273616 PMCID: PMC2686409 DOI: 10.1083/jcb.200807029] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Neuronal axons are guided by attractive and repulsive cues in their local environment. Because the repulsive guidance molecule A (RGMa) was originally identified as an axon repellent in the visual system, diverse functions in the developing and adult central nervous system have been ascribed to it. RGMa binding to its receptor neogenin induces RhoA activation, leading to inhibitory/repulsive behavior and collapse of the neuronal growth cone. However, the precise mechanisms that regulate RhoA activation are poorly understood. In this study, we show that Unc5B, a member of the netrin receptor family, interacts with neogenin as a coreceptor for RGMa. Moreover, leukemia-associated guanine nucleotide exchange factor (LARG) associates with Unc5B to transduce the RhoA signal. Focal adhesion kinase (FAK) is involved in RGMa-induced tyrosine phosphorylation of LARG as well as RhoA activation. These findings uncover the molecular basis for diverse functions mediated by RGMa.
Collapse
Affiliation(s)
- Katsuhiko Hata
- Department of Molecular Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan.
| | | | | | | |
Collapse
|
50
|
Fischer RS, Gardel M, Ma X, Adelstein RS, Waterman CM. Local cortical tension by myosin II guides 3D endothelial cell branching. Curr Biol 2009; 19:260-5. [PMID: 19185493 DOI: 10.1016/j.cub.2008.12.045] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 12/19/2008] [Accepted: 12/22/2008] [Indexed: 11/19/2022]
Abstract
A key feature of angiogenesis is directional control of endothelial cell (EC) morphogenesis and movement [1]. During angiogenic sprouting, endothelial "tip cells" directionally branch from existing vessels in response to biochemical cues such as VEGF or hypoxia and migrate and invade the surrounding extracellular matrix (ECM) in a process that requires ECM remodeling by matrix metalloproteases (MMPs) [2-4]. Tip EC branching is mediated by directional protrusion of subcellular pseudopodial branches [5, 6]. Here, we seek to understand how EC pseudopodial branching is locally regulated to directionally guide angiogenesis. We develop an in vitro 3D EC model system in which migrating ECs display branched pseudopodia morphodynamics similar to those in living zebrafish. Using this system, we find that ECM stiffness and ROCK-mediated myosin II activity inhibit EC pseudopodial branch initiation. Myosin II is dynamically localized to the EC cortex and is partially released under conditions that promote branching. Local depletion of cortical myosin II precedes branch initiation, and initiation can be induced by local inhibition of myosin II activity. Thus, local downregulation of myosin II cortical contraction allows pseudopodium initiation to mediate EC branching and hence guide directional migration and angiogenesis.
Collapse
Affiliation(s)
- Robert S Fischer
- Laboratory of Cell and Tissue Morphodynamics, Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|