1
|
Koike S, Tachikawa M, Tsutsumi M, Okada T, Nemoto T, Keino-Masu K, Masu M. Actin dynamics switches two distinct modes of endosomal fusion in yolk sac visceral endoderm cells. eLife 2024; 13:RP95999. [PMID: 39441732 PMCID: PMC11498936 DOI: 10.7554/elife.95999] [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] [Indexed: 10/25/2024] Open
Abstract
Membranes undergo various patterns of deformation during vesicle fusion, but how this membrane deformation is regulated and contributes to fusion remains unknown. In this study, we developed a new method of observing the fusion of individual late endosomes and lysosomes by using mouse yolk sac visceral endoderm cells that have huge endocytic vesicles. We found that there were two distinct fusion modes that were differently regulated. In homotypic fusion, two late endosomes fused quickly, whereas in heterotypic fusion they fused to lysosomes slowly. Mathematical modeling showed that vesicle size is a critical determinant of these fusion types and that membrane fluctuation forces can overcome the vesicle size effects. We found that actin filaments were bound to late endosomes and forces derived from dynamic actin remodeling were necessary for quick fusion during homotypic fusion. Furthermore, cofilin played a role in endocytic fusion by regulating actin turnover. These data suggest that actin promotes vesicle fusion for efficient membrane trafficking in visceral endoderm cells.
Collapse
Affiliation(s)
- Seiichi Koike
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
- Laboratory of Molecular and Cellular Biology, Graduate School of Science and Engineering for Research, University of ToyamaToyamaJapan
| | - Masashi Tachikawa
- Graduate School of Nanobioscience, Yokohama City UniversityYokohamaJapan
| | - Motosuke Tsutsumi
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural SciencesOkazakiJapan
- National Institute for Physiological Sciences, National Institutes of Natural SciencesOkazakiJapan
| | - Takuya Okada
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| | - Tomomi Nemoto
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural SciencesOkazakiJapan
- National Institute for Physiological Sciences, National Institutes of Natural SciencesOkazakiJapan
| | - Kazuko Keino-Masu
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| | - Masayuki Masu
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| |
Collapse
|
2
|
Narayanan R, Levone BR, Winterer J, Nanda P, Müller A, Lobriglio T, Fiore R, Germain PL, Mihailovich M, Testa G, Schratt G. miRNA-mediated inhibition of an actomyosin network in hippocampal pyramidal neurons restricts sociability in adult male mice. Cell Rep 2024; 43:114429. [PMID: 38968074 DOI: 10.1016/j.celrep.2024.114429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/07/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024] Open
Abstract
Social deficits are frequently observed in patients suffering from neurodevelopmental disorders, but the molecular mechanisms regulating sociability are still poorly understood. We recently reported that the loss of the microRNA (miRNA) cluster miR-379-410 leads to hypersocial behavior and anxiety in mice. Here, we show that ablating miR-379-410 in excitatory neurons of the postnatal mouse hippocampus recapitulates hypersociability, but not anxiety. At the cellular level, miR-379-410 loss in excitatory neurons leads to larger dendritic spines, increased excitatory synaptic transmission, and upregulation of an actomyosin gene network. Re-expression of three cluster miRNAs, as well as pharmacological inhibition of the actomyosin activator ROCK, is sufficient to reinstate normal sociability in miR-379-410 knockout mice. Several actomyosin genes and miR-379-410 family members are reciprocally dysregulated in isogenic human induced pluripotent stem cell (iPSC)-derived neurons harboring a deletion present in patients with Williams-Beuren syndrome, characterized by hypersocial behavior. Together, our results show an miRNA-actomyosin pathway involved in social behavior regulation.
Collapse
Affiliation(s)
- Ramanathan Narayanan
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Brunno Rocha Levone
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Jochen Winterer
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Prakruti Nanda
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Alexander Müller
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Thomas Lobriglio
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Roberto Fiore
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Pierre-Luc Germain
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland; Laboratory of Molecular and Behavioural Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland; Laboratory of Statistical Bioinformatics, IMLS, University of Zürich, Zürich, Switzerland
| | - Marija Mihailovich
- European Institute of Oncology (IEO) IRCCS, Milan, Italy; Human Technopole, Milan, Italy
| | - Giuseppe Testa
- European Institute of Oncology (IEO) IRCCS, Milan, Italy; Human Technopole, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Gerhard Schratt
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland.
| |
Collapse
|
3
|
Eberhard D, Balkenhol S, Köster A, Follert P, Upschulte E, Ostermann P, Kirschner P, Uhlemeyer C, Charnay I, Preuss C, Trenkamp S, Belgardt BF, Dickscheid T, Esposito I, Roden M, Lammert E. Semaphorin-3A regulates liver sinusoidal endothelial cell porosity and promotes hepatic steatosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:734-753. [PMID: 39196233 PMCID: PMC11358038 DOI: 10.1038/s44161-024-00487-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/07/2024] [Indexed: 08/29/2024]
Abstract
Prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease, increases worldwide and associates with type 2 diabetes and other cardiometabolic diseases. Here we demonstrate that Sema3a is elevated in liver sinusoidal endothelial cells of animal models for obesity, type 2 diabetes and MASLD. In primary human liver sinusoidal endothelial cells, saturated fatty acids induce expression of SEMA3A, and loss of a single allele is sufficient to reduce hepatic fat content in diet-induced obese mice. We show that semaphorin-3A regulates the number of fenestrae through a signaling cascade that involves neuropilin-1 and phosphorylation of cofilin-1 by LIM domain kinase 1. Finally, inducible vascular deletion of Sema3a in adult diet-induced obese mice reduces hepatic fat content and elevates very low-density lipoprotein secretion. Thus, we identified a molecular pathway linking hyperlipidemia to microvascular defenestration and early development of MASLD.
Collapse
Affiliation(s)
- Daniel Eberhard
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Metabolic Physiology, Düsseldorf, Germany
| | - Sydney Balkenhol
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Metabolic Physiology, Düsseldorf, Germany
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andrea Köster
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Metabolic Physiology, Düsseldorf, Germany
| | - Paula Follert
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Metabolic Physiology, Düsseldorf, Germany
| | - Eric Upschulte
- Cécile & Oskar Vogt Institute of Brain Research, Medical Faculty and University Hospital Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Helmholtz AI, Research Center Jülich, Jülich, Germany
| | - Philipp Ostermann
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Metabolic Physiology, Düsseldorf, Germany
| | - Philip Kirschner
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Metabolic Physiology, Düsseldorf, Germany
| | - Celina Uhlemeyer
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Iannis Charnay
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Metabolic Physiology, Düsseldorf, Germany
| | - Christina Preuss
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Sandra Trenkamp
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Bengt-Frederik Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Timo Dickscheid
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Helmholtz AI, Research Center Jülich, Jülich, Germany
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Computer Science, Düsseldorf, Germany
| | - Irene Esposito
- Institute of Pathology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Eckhard Lammert
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Metabolic Physiology, Düsseldorf, Germany.
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
4
|
Roy A, Pathak Z, Kumar H. Investigating LIM (Lin-11, Isl-1, and Mec-3) Kinases and Their Correlation with Pathological Events and Microtubule Dynamics in an Experimental Model of Spinal Cord Injury in Rats. ACS Pharmacol Transl Sci 2024; 7:667-679. [PMID: 38481685 PMCID: PMC10928889 DOI: 10.1021/acsptsci.3c00272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2025]
Abstract
The spinal cord injury (SCI) and the neurodegenerative processes accompanying it follow an intricate pathway with very limited options for treatment strategies until now. Microtubules, essential for the growth and maintenance of neurons, are mostly disorganized and destabilized due to neurodegeneration. Regeneration or plasticity is restricted to the adult central nervous system (CNS) due to several intrinsic and extrinsic mechanisms. Some fundamental or injury-induced expressions of specific molecules can be inhibited or antagonized pharmacologically to protect neurons to a certain extent after neurodegeneration. Accordingly, these molecules offer an excellent target as a therapeutic approach to promote neuroprotection. LIM kinases (LIMKs) are one of these molecules that phosphorylates members of the actin-depolymerizing factor (ADF)/cofilin family of actin-binding and filament-severing proteins. The individual role of LIMKs has not yet been studied in the pathology of SCI. In this study, we targeted LIMK and checked its role in microtubule destabilization in vitro. LIMK1 was found to be upregulated after microtubule depolymerization and inhibition of LIMK with specific inhibitor-protected neurons. Then, we checked the expressions of individual LIMKs throughout different time points across SCI in a rat contusion model, correlating with established pathophysiological markers. The phosphorylated form of LIMK1 was found to be elevated at chronic time points after injury, where scar formation and diminution of neurons prevail. Finally, we targeted the LIMK pathway with its specific inhibitor BMS-5, which showed neuroprotection after SCI. Overall, our results provided a concept concerning how a small-molecule inhibitor of LIMK may offer a strategy to treat SCI-associated neurodegeneration.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Zarna Pathak
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
5
|
Schneider F, Metz I, Rust MB. Regulation of actin filament assembly and disassembly in growth cone motility and axon guidance. Brain Res Bull 2023; 192:21-35. [PMID: 36336143 DOI: 10.1016/j.brainresbull.2022.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Directed outgrowth of axons is fundamental for the establishment of neuronal networks. Axon outgrowth is guided by growth cones, highly motile structures enriched in filamentous actin (F-actin) located at the axons' distal tips. Growth cones exploit F-actin-based protrusions to scan the environment for guidance cues, and they contain the sensory apparatus to translate guidance cue information into intracellular signaling cascades. These cascades act upstream of actin-binding proteins (ABP) and thereby control assembly and disassembly of F-actin. Spatiotemporally controlled F-actin dis-/assembly in growth cones steers the axon towards attractants and away from repellents, and it thereby navigates the axon through the developing nervous system. Hence, ABP that control F-actin dynamics emerged as critical regulators of neuronal network formation. In the present review article, we will summarize and discuss current knowledge of the mechanisms that control remodeling of the actin cytoskeleton in growth cones, focusing on recent progress in the field. Further, we will introduce tools and techniques that allow to study actin regulatory mechanism in growth cones.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Molecular Urooncology, Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany.
| |
Collapse
|
6
|
Wurz AI, Schulz AM, O’Bryant CT, Sharp JF, Hughes RM. Cytoskeletal dysregulation and neurodegenerative disease: Formation, monitoring, and inhibition of cofilin-actin rods. Front Cell Neurosci 2022; 16:982074. [PMID: 36212686 PMCID: PMC9535683 DOI: 10.3389/fncel.2022.982074] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 12/04/2022] Open
Abstract
The presence of atypical cytoskeletal dynamics, structures, and associated morphologies is a common theme uniting numerous diseases and developmental disorders. In particular, cytoskeletal dysregulation is a common cellular feature of Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. While the numerous activators and inhibitors of dysregulation present complexities for characterizing these elements as byproducts or initiators of the disease state, it is increasingly clear that a better understanding of these anomalies is critical for advancing the state of knowledge and plan of therapeutic attack. In this review, we focus on the hallmarks of cytoskeletal dysregulation that are associated with cofilin-linked actin regulation, with a particular emphasis on the formation, monitoring, and inhibition of cofilin-actin rods. We also review actin-associated proteins other than cofilin with links to cytoskeleton-associated neurodegenerative processes, recognizing that cofilin-actin rods comprise one strand of a vast web of interactions that occur as a result of cytoskeletal dysregulation. Our aim is to present a current perspective on cytoskeletal dysregulation, connecting recent developments in our understanding with emerging strategies for biosensing and biomimicry that will help shape future directions of the field.
Collapse
Affiliation(s)
- Anna I. Wurz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Anna M. Schulz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Collin T. O’Bryant
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Josephine F. Sharp
- Department of Chemistry, Notre Dame College, South Euclid, OH, United States
| | - Robert M. Hughes
- Department of Chemistry, East Carolina University, Greenville, NC, United States
- *Correspondence: Robert M. Hughes,
| |
Collapse
|
7
|
Mehrotra S, Pierce ML, Dravid SM, Murray TF. Stimulation of Neurite Outgrowth in Cerebrocortical Neurons by Sodium Channel Activator Brevetoxin-2 Requires Both N-Methyl-D-aspartate Receptor 2B (GluN2B) and p21 Protein (Cdc42/Rac)-Activated Kinase 1 (PAK1). Mar Drugs 2022; 20:559. [PMID: 36135748 PMCID: PMC9504648 DOI: 10.3390/md20090559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 12/05/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors play a critical role in activity-dependent dendritic arborization, spinogenesis, and synapse formation by stimulating calcium-dependent signaling pathways. Previously, we have shown that brevetoxin 2 (PbTx-2), a voltage-gated sodium channel (VGSC) activator, produces a concentration-dependent increase in intracellular sodium [Na+]I and increases NMDA receptor (NMDAR) open probabilities and NMDA-induced calcium (Ca2+) influxes. The objective of this study is to elucidate the downstream signaling mechanisms by which the sodium channel activator PbTx-2 influences neuronal morphology in murine cerebrocortical neurons. PbTx-2 and NMDA triggered distinct Ca2+-influx pathways, both of which involved the NMDA receptor 2B (GluN2B). PbTx-2-induced neurite outgrowth in day in vitro 1 (DIV-1) neurons required the small Rho GTPase Rac1 and was inhibited by both a PAK1 inhibitor and a PAK1 siRNA. PbTx-2 exposure increased the phosphorylation of PAK1 at Thr-212. At DIV-5, PbTx-2 induced increases in dendritic protrusion density, p-cofilin levels, and F-actin throughout the dendritic arbor and soma. Moreover, PbTx-2 increased miniature excitatory post-synaptic currents (mEPSCs). These data suggest that the stimulation of neurite outgrowth, spinogenesis, and synapse formation produced by PbTx-2 are mediated by GluN2B and PAK1 signaling.
Collapse
Affiliation(s)
- Suneet Mehrotra
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
- Omeros, Seattle, WA 98119, USA
| | - Marsha L. Pierce
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
- Department of Pharmacology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Shashank M. Dravid
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Thomas F. Murray
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| |
Collapse
|
8
|
Lai YJ, Tsai FC, Chang GJ, Chang SH, Huang CC, Chen WJ, Yeh YH. miR-181b targets semaphorin 3A to mediate TGF-β-induced endothelial-mesenchymal transition related to atrial fibrillation. J Clin Invest 2022; 132:142548. [PMID: 35775491 PMCID: PMC9246393 DOI: 10.1172/jci142548] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Atrial fibrosis is an essential contributor to atrial fibrillation (AF). It remains unclear whether atrial endocardial endothelial cells (AEECs) that undergo endothelial-mesenchymal transition (EndMT) are among the sources of atrial fibroblasts. We studied human atria, TGF-β-treated human AEECs, cardiac-specific TGF-β-transgenic mice, and heart failure rabbits to identify the underlying mechanism of EndMT in atrial fibrosis. Using isolated AEECs, we found that miR-181b was induced in TGF-β-treated AEECs, which decreased semaphorin 3A (Sema3A) and increased EndMT markers, and these effects could be reversed by a miR-181b antagomir. Experiments in which Sema3A was increased by a peptide or decreased by a siRNA in AEECs revealed a mechanistic link between Sema3A and LIM-kinase 1/phosphorylated cofilin (LIMK/p-cofilin) signaling and suggested that Sema3A is upstream of LIMK in regulating actin remodeling through p-cofilin. Administration of the miR-181b antagomir or recombinant Sema3A to TGF-β-transgenic mice evoked increased Sema3A, reduced EndMT markers, and significantly decreased atrial fibrosis and AF vulnerability. Our study provides a mechanistic link between the induction of EndMT by TGF-β via miR-181b/Sema3A/LIMK/p-cofilin signaling to atrial fibrosis. Blocking miR-181b and increasing Sema3A are potential strategies for AF therapeutic intervention.
Collapse
Affiliation(s)
- Ying-Ju Lai
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao Yuan, Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chia Yi, Taiwan
| | - Feng-Chun Tsai
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Gwo-Jyh Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Tao Yuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Chung-Chi Huang
- Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao Yuan, Taiwan.,Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Tao Yuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| |
Collapse
|
9
|
Ribba AS, Fraboulet S, Sadoul K, Lafanechère L. The Role of LIM Kinases during Development: A Lens to Get a Glimpse of Their Implication in Pathologies. Cells 2022; 11:cells11030403. [PMID: 35159213 PMCID: PMC8834001 DOI: 10.3390/cells11030403] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/24/2022] Open
Abstract
The organization of cell populations within animal tissues is essential for the morphogenesis of organs during development. Cells recognize three-dimensional positions with respect to the whole organism and regulate their cell shape, motility, migration, polarization, growth, differentiation, gene expression and cell death according to extracellular signals. Remodeling of the actin filaments is essential to achieve these cell morphological changes. Cofilin is an important binding protein for these filaments; it increases their elasticity in terms of flexion and torsion and also severs them. The activity of cofilin is spatiotemporally inhibited via phosphorylation by the LIM domain kinases 1 and 2 (LIMK1 and LIMK2). Phylogenetic analysis indicates that the phospho-regulation of cofilin has evolved as a mechanism controlling the reorganization of the actin cytoskeleton during complex multicellular processes, such as those that occur during embryogenesis. In this context, the main objective of this review is to provide an update of the respective role of each of the LIM kinases during embryonic development.
Collapse
|
10
|
Creighton BA, Afriyie S, Ajit D, Casingal CR, Voos KM, Reger J, Burch AM, Dyne E, Bay J, Huang JK, Anton ES, Fu MM, Lorenzo DN. Giant ankyrin-B mediates transduction of axon guidance and collateral branch pruning factor sema 3A. eLife 2021; 10:69815. [PMID: 34812142 PMCID: PMC8610419 DOI: 10.7554/elife.69815] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 11/04/2021] [Indexed: 01/19/2023] Open
Abstract
Variants in the high confident autism spectrum disorder (ASD) gene ANK2 target both ubiquitously expressed 220 kDa ankyrin-B and neurospecific 440 kDa ankyrin-B (AnkB440) isoforms. Previous work showed that knock-in mice expressing an ASD-linked Ank2 variant yielding a truncated AnkB440 product exhibit ectopic brain connectivity and behavioral abnormalities. Expression of this variant or loss of AnkB440 caused axonal hyperbranching in vitro, which implicated AnkB440 microtubule bundling activity in suppressing collateral branch formation. Leveraging multiple mouse models, cellular assays, and live microscopy, we show that AnkB440 also modulates axon collateral branching stochastically by reducing the number of F-actin-rich branch initiation points. Additionally, we show that AnkB440 enables growth cone (GC) collapse in response to chemorepellent factor semaphorin 3 A (Sema 3 A) by stabilizing its receptor complex L1 cell adhesion molecule/neuropilin-1. ASD-linked ANK2 variants failed to rescue Sema 3A-induced GC collapse. We propose that impaired response to repellent cues due to AnkB440 deficits leads to axonal targeting and branch pruning defects and may contribute to the pathogenicity of ANK2 variants.
Collapse
Affiliation(s)
- Blake A Creighton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Simone Afriyie
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Deepa Ajit
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Cristine R Casingal
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Kayleigh M Voos
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Joan Reger
- National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, United States.,Department of Biology and Center for Cell Reprogramming, Georgetown University, Washington, United States
| | - April M Burch
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Eric Dyne
- National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, United States
| | - Julia Bay
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Jeffrey K Huang
- Department of Biology and Center for Cell Reprogramming, Georgetown University, Washington, United States
| | - E S Anton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Meng-Meng Fu
- National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, United States
| | - Damaris N Lorenzo
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Carolina Institute for Developmental Disabilities, Chapel Hill, United States
| |
Collapse
|
11
|
Namme JN, Bepari AK, Takebayashi H. Cofilin Signaling in the CNS Physiology and Neurodegeneration. Int J Mol Sci 2021; 22:ijms221910727. [PMID: 34639067 PMCID: PMC8509315 DOI: 10.3390/ijms221910727] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/26/2021] [Accepted: 09/29/2021] [Indexed: 12/18/2022] Open
Abstract
All eukaryotic cells are composed of the cytoskeleton, which plays crucial roles in coordinating diverse cellular functions such as cell division, morphology, migration, macromolecular stabilization, and protein trafficking. The cytoskeleton consists of microtubules, intermediate filaments, and actin filaments. Cofilin, an actin-depolymerizing protein, is indispensable for regulating actin dynamics in the central nervous system (CNS) development and function. Cofilin activities are spatiotemporally orchestrated by numerous extra- and intra-cellular factors. Phosphorylation at Ser-3 by kinases attenuate cofilin’s actin-binding activity. In contrast, dephosphorylation at Ser-3 enhances cofilin-induced actin depolymerization. Cofilin functions are also modulated by various binding partners or reactive oxygen species. Although the mechanism of cofilin-mediated actin dynamics has been known for decades, recent research works are unveiling the profound impacts of cofilin dysregulation in neurodegenerative pathophysiology. For instance, oxidative stress-induced increase in cofilin dephosphorylation is linked to the accumulation of tau tangles and amyloid-beta plaques in Alzheimer’s disease. In Parkinson’s disease, cofilin activation by silencing its upstream kinases increases α-synuclein-fibril entry into the cell. This review describes the molecular mechanism of cofilin-mediated actin dynamics and provides an overview of cofilin’s importance in CNS physiology and pathophysiology.
Collapse
Affiliation(s)
- Jannatun Nayem Namme
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh;
| | - Asim Kumar Bepari
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh;
- Correspondence: (A.K.B.); (H.T.)
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
- Correspondence: (A.K.B.); (H.T.)
| |
Collapse
|
12
|
Hou X, Nozumi M, Nakamura H, Igarashi M, Sugiyama S. Coactosin Promotes F-Actin Protrusion in Growth Cones Under Cofilin-Related Signaling Pathway. Front Cell Dev Biol 2021; 9:660349. [PMID: 34235144 PMCID: PMC8256272 DOI: 10.3389/fcell.2021.660349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/31/2021] [Indexed: 12/03/2022] Open
Abstract
During brain development, axon outgrowth and its subsequent pathfinding are reliant on a highly motile growth cone located at the tip of the axon. Actin polymerization that is regulated by actin-depolymerizing factors homology (ADF-H) domain-containing family drives the formation of lamellipodia and filopodia at the leading edge of growth cones for axon guidance. However, the precise localization and function of ADF-H domain-containing proteins involved in axon extension and retraction remain unclear. We have previously shown that transcripts and proteins of coactosin-like protein 1 (COTL1), an ADF-H domain-containing protein, are observed in neurites and axons in chick embryos. Coactosin overexpression analysis revealed that this protein was localized to axonal growth cones and involved in axon extension in the midbrain. We further examined the specific distribution of coactosin and cofilin within the growth cone using superresolution microscopy, structured illumination microscopy, which overcomes the optical diffraction limitation and is suitable to the analysis of cellular dynamic movements. We found that coactosin was tightly associated with F-actin bundles at the growth cones and that coactosin overexpression promoted the expansion of lamellipodia and extension of growth cones. Coactosin knockdown in oculomotor neurons resulted in an increase in the levels of the inactive, phosphorylated form of cofilin and dysregulation of actin polymerization and axonal elongation, which suggests that coactosin promoted axonal growth in a cofilin-dependent manner. Indeed, the application of a dominant-negative form of LIMK1, a downstream effector of GTPases, reversed the effect of coactosin knockdown on axonal growth by enhancing cofilin activity. Combined, our results indicate that coactosin functions promote the assembly of protrusive actin filament arrays at the leading edge for growth cone motility.
Collapse
Affiliation(s)
- Xubin Hou
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Department of Molecular Neurobiology, Graduate School of Life Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Harukazu Nakamura
- Department of Molecular Neurobiology, Graduate School of Life Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Sayaka Sugiyama
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
13
|
Henneberger C, Bard L, Panatier A, Reynolds JP, Kopach O, Medvedev NI, Minge D, Herde MK, Anders S, Kraev I, Heller JP, Rama S, Zheng K, Jensen TP, Sanchez-Romero I, Jackson CJ, Janovjak H, Ottersen OP, Nagelhus EA, Oliet SHR, Stewart MG, Nägerl UV, Rusakov DA. LTP Induction Boosts Glutamate Spillover by Driving Withdrawal of Perisynaptic Astroglia. Neuron 2020; 108:919-936.e11. [PMID: 32976770 PMCID: PMC7736499 DOI: 10.1016/j.neuron.2020.08.030] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 06/14/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023]
Abstract
Extrasynaptic actions of glutamate are limited by high-affinity transporters expressed by perisynaptic astroglial processes (PAPs): this helps maintain point-to-point transmission in excitatory circuits. Memory formation in the brain is associated with synaptic remodeling, but how this affects PAPs and therefore extrasynaptic glutamate actions is poorly understood. Here, we used advanced imaging methods, in situ and in vivo, to find that a classical synaptic memory mechanism, long-term potentiation (LTP), triggers withdrawal of PAPs from potentiated synapses. Optical glutamate sensors combined with patch-clamp and 3D molecular localization reveal that LTP induction thus prompts spatial retreat of astroglial glutamate transporters, boosting glutamate spillover and NMDA-receptor-mediated inter-synaptic cross-talk. The LTP-triggered PAP withdrawal involves NKCC1 transporters and the actin-controlling protein cofilin but does not depend on major Ca2+-dependent cascades in astrocytes. We have therefore uncovered a mechanism by which a memory trace at one synapse could alter signal handling by multiple neighboring connections.
Collapse
Affiliation(s)
- Christian Henneberger
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK; Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany.
| | - Lucie Bard
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Aude Panatier
- INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France; Université de Bordeaux, 33000 Bordeaux, France; Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - James P Reynolds
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Olga Kopach
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | | | - Daniel Minge
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Michel K Herde
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Stefanie Anders
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Igor Kraev
- Life Sciences, The Open University, Milton Keynes MK7 6AA, UK
| | - Janosch P Heller
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sylvain Rama
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Kaiyu Zheng
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Thomas P Jensen
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | | | - Colin J Jackson
- Research School of Chemistry, Australian National University, Acton, ACT 2601, Australia
| | - Harald Janovjak
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria; EMBL Australia, Australian Regenerative Medicine Institute, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, VIC 3800, Australia
| | - Ole Petter Ottersen
- Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Stephane H R Oliet
- INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France; Université de Bordeaux, 33000 Bordeaux, France
| | | | - U Valentin Nägerl
- Université de Bordeaux, 33000 Bordeaux, France; Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France.
| | - Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK.
| |
Collapse
|
14
|
Tian F, Wang X, Ni H, Feng X, Yuan X, Huang Q. The ginsenoside metabolite compound K stimulates glucagon-like peptide-1 secretion in NCI-H716 cells by regulating the RhoA/ROCKs/YAP signaling pathway and cytoskeleton formation. J Pharmacol Sci 2020; 145:88-96. [PMID: 33357784 DOI: 10.1016/j.jphs.2020.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 02/08/2023] Open
Abstract
Ginsenoside Rb1 has been shown to have antidiabetic and anti-inflammatory effects. Its major metabolite, compound K (CK), can stimulate the secretion of glucagon-like peptide-1 (GLP1), a gastrointestinal hormone that plays a vital role in regulating glucose metabolism. However, the mechanism underlying the regulation of GLP1 secretion by compound K has not been fully explored. This study was designed to investigate whether CK ameliorates incretin impairment by regulating the RhoA/ROCKs/YAP signaling pathway and cytoskeleton formation in NCI-H716 cells. Using NCI-H716 cells as a model cell line for GLP1 secretion, we analyzed the effect of CK on the expression of RhoA/ROCK/YAP pathway components. Our results suggest that the effect of CK on GLP1 secretion depends on the anti-inflammatory effect of CK. We also demonstrated that CK can affect the RhoA/ROCK/YAP pathway, which is downstream of transforming growth factor β1 (TGFβ1), by maintaining the capacity of intestinal differentiation. In addition, this effect was mediated by regulating F/G-actin dynamics. These results provide not only the mechanistic insight for the effect of CK on intestinal L cells but also the molecular basis for the further development of CK as a potential therapeutic agent to treat type 2 diabetes mellitus (T2D).
Collapse
Affiliation(s)
- Fengyuan Tian
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| | - Xi Wang
- Central Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, PR China.
| | - Haixiang Ni
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| | - Xiaohong Feng
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| | - Xiao Yuan
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| | - Qi Huang
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| |
Collapse
|
15
|
Faini G, Del Bene F, Albadri S. Reelin functions beyond neuronal migration: from synaptogenesis to network activity modulation. Curr Opin Neurobiol 2020; 66:135-143. [PMID: 33197872 DOI: 10.1016/j.conb.2020.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/08/2020] [Accepted: 10/11/2020] [Indexed: 01/05/2023]
Abstract
Reelin, a glycoprotein of the extracellular matrix, has been the focus of several studies over the years, mostly for its role in cell migration. Here we report the role of this molecule and of its downstream pathways in post-mitotic neurons and how they contribute to neural circuit assembly, refinement and function. Accumulating evidence has pointed at a major role for Reelin in axonal guidance, synaptogenesis and dendritic spine formation. In particular, new evidence points at a direct role in axonal targeting and refinement at the target site. In addition, recent advances highlight new functions of Reelin in the modulation of synaptic activity, plasticity and behavior and in the direct regulation of GABA receptors expression and stability. We discuss these findings in the context of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Giulia Faini
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Filippo Del Bene
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.
| | - Shahad Albadri
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
16
|
Reelin-Nrp1 Interaction Regulates Neocortical Dendrite Development in a Context-Specific Manner. J Neurosci 2020; 40:8248-8261. [PMID: 33009002 DOI: 10.1523/jneurosci.1907-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 11/21/2022] Open
Abstract
Reelin plays versatile roles in neocortical development. The C-terminal region (CTR) of Reelin is required for the correct formation of the superficial structure of the neocortex; however, the mechanisms by which this position-specific effect occurs remain largely unknown. In this study, we demonstrate that Reelin with an intact CTR binds to neuropilin-1 (Nrp1), a transmembrane protein. Both male and female mice were used. Nrp1 is localized with very-low-density lipoprotein receptor (VLDLR), a canonical Reelin receptor, in the superficial layers of the developing neocortex. It forms a complex with VLDLR, and this interaction is modulated by the alternative splicing of VLDLR. Reelin with an intact CTR binds more strongly to the VLDLR/Nrp1 complex than to VLDLR alone. Knockdown of Nrp1 in neurons leads to the accumulation of Dab1 protein. Since the degradation of Dab1 is induced by Reelin signaling, it is suggested that Nrp1 augments Reelin signaling. The interaction between Reelin and Nrp1 is required for normal dendritic development in superficial-layer neurons. All of these characteristics of Reelin are abrogated by proteolytic processing of the six C-terminal amino acid residues of Reelin (0.17% of the whole protein). Therefore, Nrp1 is a coreceptor molecule for Reelin and, together with the proteolytic processing of Reelin, can account for context-specific Reelin function in brain development.SIGNIFICANCE STATEMENT Reelin often exhibits a context-dependent function during brain development; however, its underlying mechanism is not well understood. We found that neuropilin-1 (Nrp1) specifically binds to the CTR of Reelin and acts as a coreceptor for very-low-density lipoprotein receptor (VLDLR). The Nrp1/VLDLR complex is localized in the superficial layers of the neocortex, and its interaction with Reelin is essential for proper dendritic development in superficial-layer neurons. This study provides the first mechanistic evidence of the context-specific function of Reelin (>3400 residues) regulated by the C-terminal residues and Nrp1, a component of the canonical Reelin receptor complex.
Collapse
|
17
|
Wang Y, Song X, Wang Y, Huang L, Luo W, Li F, Qin S, Wang Y, Xiao J, Wu Y, Jin F, Kitazato K, Wang Y. Dysregulation of cofilin-1 activity-the missing link between herpes simplex virus type-1 infection and Alzheimer's disease. Crit Rev Microbiol 2020; 46:381-396. [PMID: 32715819 DOI: 10.1080/1040841x.2020.1794789] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial disease triggered by environmental factors in combination with genetic predisposition. Infectious agents, in particular herpes simplex virus type 1 (HSV-1), are gradually being recognised as important factors affecting the development of AD. However, the mechanism linking HSV-1 and AD remains unknown. Of note, HSV-1 manipulates the activity of cofilin-1 to ensure their efficient infection in neuron cells. Cofilin-1, the main regulator of actin cytoskeleton reorganization, is implicating for the plastic of dendritic spines and axon regeneration of neuronal cells. Moreover, dysfunction of cofilin-1 is observed in most AD patients, as well as in mice with AD and ageing. Further, inhibition of cofilin-1 activity ameliorates the host cognitive impairment in an animal model of AD. Together, dysregulation of cofilin-1 led by HSV-1 infection is a potential link between HSV-1 and AD. Herein, we critically summarize the role of cofilin-1-mediated actin dynamics in both HSV-1 infection and AD, respectively. We also propose several hypotheses regarding the connecting roles of cofilin-1 dysregulation in HSV-1 infection and AD. Our review provides a foundation for future studies targeting individuals carrying HSV-1 in combination with cofilin-1 to promote a more individualised approach for treatment and prevention of AD.
Collapse
Affiliation(s)
- Yiliang Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Xiaowei Song
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Yun Wang
- Department of Obstetrics and gynecology, The First affiliated hospital of Jinan University, Guangzhou, PR China
| | - Lianzhou Huang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Weisheng Luo
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Feng Li
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Shurong Qin
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Yuan Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Ji Xiao
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Yanting Wu
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Fujun Jin
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| | - Kaio Kitazato
- Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yifei Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, PR China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, PR China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, PR China
| |
Collapse
|
18
|
Ikeno T, Konishi Y. Arp2/3 Is Required for Axonal Arbor Terminal Retraction in Cerebellar Granule Neurons. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420010109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Takabatake M, Goshima Y, Sasaki Y. Semaphorin-3A Promotes Degradation of Fragile X Mental Retardation Protein in Growth Cones via the Ubiquitin-Proteasome Pathway. Front Neural Circuits 2020; 14:5. [PMID: 32184710 PMCID: PMC7059091 DOI: 10.3389/fncir.2020.00005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/07/2020] [Indexed: 01/07/2023] Open
Abstract
Fragile X mental retardation protein (FMRP) is an RNA-binding protein that regulates local translation in dendrites and spines for synaptic plasticity. In axons, FMRP is implicated in axonal extension and axon guidance. We previously demonstrated the involvement of FMRP in growth cone collapse via a translation-dependent response to Semaphorin-3A (Sema3A), a repulsive axon guidance factor. In the case of attractive axon guidance factors, RNA-binding proteins such as zipcode binding protein 1 (ZBP1) accumulate towards the stimulated side of growth cones for local translation. However, it remains unclear how Sema3A effects FMRP localization in growth cones. Here, we show that levels of FMRP in growth cones of hippocampal neurons decreased after Sema3A stimulation. This decrease in FMRP was suppressed by the ubiquitin-activating enzyme E1 enzyme inhibitor PYR-41 and proteasome inhibitor MG132, suggesting that the ubiquitin-proteasome pathway is involved in Sema3A-induced FMRP degradation in growth cones. Moreover, the E1 enzyme or proteasome inhibitor suppressed Sema3A-induced increases in microtubule-associated protein 1B (MAP1B) in growth cones, suggesting that the ubiquitin-proteasome pathway promotes local translation of MAP1B, whose translation is mediated by FMRP. These inhibitors also blocked the Sema3A-induced growth cone collapse. Collectively, our results suggest that Sema3A promotes degradation of FMRP in growth cones through the ubiquitin-proteasome pathway, leading to growth cone collapse via local translation of MAP1B. These findings reveal a new mechanism of axon guidance regulation: degradation of the translational suppressor FMRP via the ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Masaru Takabatake
- Functional Structure Biology Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yukio Sasaki
- Functional Structure Biology Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| |
Collapse
|
20
|
The C. elegans BRCA2-ALP/Enigma Complex Regulates Axon Regeneration via a Rho GTPase-ROCK-MLC Phosphorylation Pathway. Cell Rep 2019; 24:1880-1889. [PMID: 30110643 DOI: 10.1016/j.celrep.2018.07.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 05/28/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Abstract
The ability of specific neurons to regenerate their axons after injury is governed by cell-intrinsic regeneration pathways. However, the mechanisms regulating axon regeneration are not well understood. Here, we identify the brc-2 gene encoding a homolog of the mammalian BRCA2 tumor suppressor as a regulator of axon regeneration in Caenorhabditis elegans motor neurons. We show that the RHO-1/Rho GTPase-LET-502/ROCK (Rho-associated coiled-coil kinase)-regulatory non-muscle myosin light-chain (MLC-4/MLC) phosphorylation signaling pathway regulates axon regeneration. BRC-2 functions between RHO-1 and LET-502, suggesting that BRC-2 is required for the activation of LET-502 by RHO-1-GTP. We also find that one component that interacts with BRC-2, the ALP (α-actinin-associated LIM protein)/Enigma protein ALP-1, is required for regeneration and acts between LET-502 and MLC-4 phosphorylation. Furthermore, we demonstrate that ALP-1 associates with LET-502 and MLC-4. Thus, ALP-1 serves as a platform to activate MLC-4 phosphorylation mediated by the RHO-1-LET-502 signaling pathway.
Collapse
|
21
|
Optogenetic control of cofilin and αTAT in living cells using Z-lock. Nat Chem Biol 2019; 15:1183-1190. [PMID: 31740825 PMCID: PMC6873228 DOI: 10.1038/s41589-019-0405-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/09/2019] [Indexed: 11/29/2022]
Abstract
Here we introduce Z-lock, an optogenetic approach for reversible, light-controlled steric inhibition of protein active sites. The LOV domain and Zdk, a small protein that binds LOV selectively in the dark, are appended to the protein of interest where they sterically block the active site. Irradiation causes LOV to change conformation and release Zdk, exposing the active site. Computer-assisted protein design was used to optimize linkers and Zdk-LOV affinity, for both effective binding in the dark, and effective light-induced release of the intramolecular interaction. Z-lock cofilin was shown to have actin severing ability in vitro, and in living cancer cells it produced protrusions and invadopodia. An active fragment of the tubulin acetylase αTAT was similarly modified and shown to acetylate tubulin upon irradiation.
Collapse
|
22
|
Wang W, Halasz E, Townes-Anderson E. Actin Dynamics, Regulated by RhoA-LIMK-Cofilin Signaling, Mediates Rod Photoreceptor Axonal Retraction After Retinal Injury. Invest Ophthalmol Vis Sci 2019; 60:2274-2285. [PMID: 31112612 PMCID: PMC6530517 DOI: 10.1167/iovs.18-26077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Purpose Retraction of the axon terminals of rod photoreceptors after retinal detachment breaks the first synapse in the visual pathway, resulting in visual impairment. Previous work showed that the mechanism of axonal retraction involves RhoA signaling and its downstream effector LIM Kinase (LIMK) activation. We examined the response of the downstream component cofilin, a direct binding protein of actin filaments, as well as the regulation by RhoA-LIMK-Cofilin signaling of actin assembly/disassembly, in the presynaptic ribbon terminal of injured rod cells. Methods Injury was produced by retinal detachment or rod cell isolation. Detached porcine retina was probed for levels and localization of phosphorylated cofilin with Western blots and confocal microscopy, whereas rod cell cultures of dissociated salamander retina were examined for filamentous actin assembly/disassembly with a barbed end assay and phalloidin staining. Results A detachment increased phosphorylation of cofilin in retinal explants; phosphorylation occurred in rod terminals in sections of detached retina. Isolation of rod cells resulted in axon retraction accompanied by an increase in actin barbed ends and a decrease in net filament labeling. All changes were significantly reduced by either Rho kinase (ROCK) or LIMK inhibition, using Y27632 or BMS-5, respectively. Cytochalasin D also reduced retraction and stabilized filaments in isolated rod cells. Conclusions These results indicate that actin depolymerization via activation of RhoA downstream kinases and cofilin contributes to axon retraction. Preventing depolymerization, in addition to actomyosin contraction, may stabilize ribbon synapses after trauma.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| | - Eva Halasz
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| | - Ellen Townes-Anderson
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| |
Collapse
|
23
|
The Cofilin/Limk1 Pathway Controls the Growth Rate of Both Developing and Regenerating Motor Axons. J Neurosci 2019; 39:9316-9327. [PMID: 31578231 DOI: 10.1523/jneurosci.0648-19.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/12/2019] [Accepted: 08/29/2019] [Indexed: 12/31/2022] Open
Abstract
Regenerating axons often have to grow considerable distances to reestablish circuits, making functional recovery a lengthy process. One solution to this problem would be to co-opt the "temporal" guidance mechanisms that control the rate of axon growth during development to accelerate the rate at which nerves regenerate in adults. We have previously found that the loss of Limk1, a negative regulator of cofilin, accelerates the rate of spinal commissural axon growth. Here, we use mouse models to show that spinal motor axon outgrowth is similarly promoted by the loss of Limk1, suggesting that temporal guidance mechanisms are widely used during development. Furthermore, we find that the regulation of cofilin activity is an acute response to nerve injury in the peripheral nervous system. Within hours of a sciatic nerve injury, the level of phosphorylated cofilin dramatically increases at the lesion site, in a Limk1-dependent manner. This response may be a major constraint on the rate of peripheral nerve regeneration. Proof-of-principle experiments show that elevating cofilin activity, through the loss of Limk1, results in faster sciatic nerve growth, and improved recovery of some sensory and motor function.SIGNIFICANCE STATEMENT The studies shed light on an endogenous, shared mechanism that controls the rate at which developing and regenerating axons grow. An understanding of these mechanisms is key for developing therapies to reduce painful recovery times for nerve-injury patients, by accelerating the rate at which damaged nerves reconnect with their synaptic targets.
Collapse
|
24
|
6-Substituted purines as ROCK inhibitors with anti-metastatic activity. Bioorg Chem 2019; 90:103005. [PMID: 31271944 DOI: 10.1016/j.bioorg.2019.103005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/31/2019] [Accepted: 05/19/2019] [Indexed: 12/12/2022]
Abstract
Rho-associated serine/threonine kinases (ROCKs) are principal regulators of the actin cytoskeleton that regulate the contractility, shape, motility, and invasion of cells. We explored the relationships between structure and anti-ROCK2 activity in a group of purine derivatives substituted at the C6 atom by piperidin-1-yl or azepan-1-yl groups. Structure-activity relationship (SAR) analyses suggested that anti-ROCK activity is retained, and may be further increased, by substitution of the parent compounds at the C2 atom or by expansion of the C6 side chain. These inhibitors of ROCK can reach effective concentrations within cells, as demonstrated by a decrease in phosphorylation of the ROCK target MLC, and by inhibition of the ROCK-dependent invasion of melanoma cells in the collagen matrix. Our study may be useful for further optimization of C6-substituted purine inhibitors of ROCKs and of other sensitive kinases identified by the screening of a broad panel of protein kinases.
Collapse
|
25
|
Ye X, Qiu Y, Gao Y, Wan D, Zhu H. A Subtle Network Mediating Axon Guidance: Intrinsic Dynamic Structure of Growth Cone, Attractive and Repulsive Molecular Cues, and the Intermediate Role of Signaling Pathways. Neural Plast 2019; 2019:1719829. [PMID: 31097955 PMCID: PMC6487106 DOI: 10.1155/2019/1719829] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 01/01/2023] Open
Abstract
A fundamental feature of both early nervous system development and axon regeneration is the guidance of axonal projections to their targets in order to assemble neural circuits that control behavior. In the navigation process where the nerves grow toward their targets, the growth cones, which locate at the tips of axons, sense the environment surrounding them, including varies of attractive or repulsive molecular cues, then make directional decisions to adjust their navigation journey. The turning ability of a growth cone largely depends on its highly dynamic skeleton, where actin filaments and microtubules play a very important role in its motility. In this review, we summarize some possible mechanisms underlying growth cone motility, relevant molecular cues, and signaling pathways in axon guidance of previous studies and discuss some questions regarding directions for further studies.
Collapse
Affiliation(s)
- Xiyue Ye
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yan Qiu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yuqing Gao
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Dong Wan
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Huifeng Zhu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| |
Collapse
|
26
|
hnRNP Q Regulates Internal Ribosome Entry Site-Mediated fmr1 Translation in Neurons. Mol Cell Biol 2019; 39:MCB.00371-18. [PMID: 30478144 DOI: 10.1128/mcb.00371-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 11/15/2018] [Indexed: 01/05/2023] Open
Abstract
Fragile X syndrome (FXS) caused by loss of fragile X mental retardation protein (FMRP), is the most common cause of inherited intellectual disability. Numerous studies show that FMRP is an RNA binding protein that regulates translation of its binding targets and plays key roles in neuronal functions. However, the regulatory mechanism for FMRP expression is incompletely understood. Conflicting results regarding internal ribosome entry site (IRES)-mediated fmr1 translation have been reported. Here, we unambiguously demonstrate that the fmr1 gene, which encodes FMRP, exploits both IRES-mediated translation and canonical cap-dependent translation. Furthermore, we find that heterogeneous nuclear ribonucleoprotein Q (hnRNP Q) acts as an IRES-transacting factor (ITAF) for IRES-mediated fmr1 translation in neurons. We also show that semaphorin 3A (Sema3A)-induced axonal growth cone collapse is due to upregulation of hnRNP Q and subsequent IRES-mediated expression of FMRP. These data elucidate the regulatory mechanism of FMRP expression and its role in axonal growth cone collapse.
Collapse
|
27
|
C-terminal fragments of amyloid precursor proteins increase cofilin phosphorylation by LIM kinase in cultured rat primary neurons. Neuroreport 2019; 30:38-45. [PMID: 30444792 DOI: 10.1097/wnr.0000000000001162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Amyloid precursor proteins (APPs) are processed by β-, γ-, and ε-secretases and caspase-3 to generate C-terminal fragments of APP (APP-CTFs), which may contribute to the pathology of Alzheimer's disease (AD). In addition to amyloid plaques and neurofibrillary tangles, AD brains contain Hirano bodies, which are rod-like structures mostly composed of actin and the actin-binding protein, cofilin. However, the mechanisms underlying the formation of cofilin-actin rods are still unknown. In this study, we aim to elucidate the effects of APP-CTFs on the actin-depolymerizing factor [(ADF)/cofilin]. Our data indicate that transfection with APP-CT99 and APP-CT57 may increase the phosphorylation level of Ser3 of ADF/cofilin and Thr508 of LIM-kinase 1 in rat primary cortical neuronal cultures. S3 peptide, a synthetic peptide competitor of LIM-kinase 1 for ADF/cofilin phosphorylation and an inhibitor of APP-CTFs, induced ADF/cofilin phosphorylation. In comparison with the wild-type mouse, the APP-CT transgenic mouse showed increased immunoreactivity of phosphorylated cofilin (p-cofilin) in the brain. Treatment with DAPT, an inhibitor of γ-secretase, resulted in a decrease in p-cofilin protein level in the group transfected with full-length APP-695. Transfection with the mutant APP-CTF with a deleted YENPTY domain resulted in no significant increase in p-cofilin level. Thus, APP-CTFs induced cofilin phosphorylation to facilitate nuclear translocation. These results suggest a relationship between APP-CTFs and ADF/cofilin that may be suggestive of a new toxic pathway in the pathology of AD.
Collapse
|
28
|
Tastet J, Cuberos H, Vallée B, Toutain A, Raynaud M, Marouillat S, Thépault RA, Laumonnier F, Bonnet-Brilhault F, Vourc'h P, Andres CR, Bénédetti H. LIMK2-1 is a Hominidae-Specific Isoform of LIMK2 Expressed in Central Nervous System and Associated with Intellectual Disability. Neuroscience 2018; 399:199-210. [PMID: 30594563 DOI: 10.1016/j.neuroscience.2018.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/24/2022]
Abstract
LIMK2 is involved in neuronal functions by regulating actin dynamics. Different isoforms of LIMK2 are described in databanks. LIMK2a and LIMK2b are the most characterized. A few pieces of evidence suggest that LIMK2 isoforms might not have overlapping functions. In this study, we focused our attention on a less studied human LIMK2 isoform, LIMK2-1. Compared to the other LIMK2 isoforms, LIMK2-1 contains a supplementary C-terminal phosphatase 1 inhibitory domain (PP1i). We found out that this isoform was hominidae-specific and showed that it was expressed in human fetal brain and faintly in adult brain. Its coding sequence was sequenced in 173 patients with sporadic non-syndromic intellectual disability (ID), and we observed an association of a rare missense variant in the PP1i domain (rs151191437, p.S668P) with ID. Our results also suggest an implication of LIMK2-1 in neurite outgrowth and neurons arborization which appears to be affected by the p.S668P variation. Therefore our results suggest that LIMK2-1 plays a role in the developing brain, and that a rare variation of this isoform is a susceptibility factor in ID.
Collapse
Affiliation(s)
- Julie Tastet
- UMR INSERM U1253, Université François Rabelais, Tours, France; CNRS UPR 4301, CBM, Orléans, France; Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Hélène Cuberos
- UMR INSERM U1253, Université François Rabelais, Tours, France; CNRS UPR 4301, CBM, Orléans, France
| | | | - Annick Toutain
- UMR INSERM U1253, Université François Rabelais, Tours, France; CHRU de Tours, Service de Génétique, Tours, France
| | - Martine Raynaud
- UMR INSERM U1253, Université François Rabelais, Tours, France; CHRU de Tours, Service de Génétique, Tours, France
| | | | | | | | - Frédérique Bonnet-Brilhault
- UMR INSERM U1253, Université François Rabelais, Tours, France; CHRU de Tours, Service de Pédopsychiatrie, Tours, France
| | - Patrick Vourc'h
- UMR INSERM U1253, Université François Rabelais, Tours, France; CHRU de Tours, Service de Biochimie et de Biologie Moléculaire, Tours, France
| | - Christian R Andres
- UMR INSERM U1253, Université François Rabelais, Tours, France; CHRU de Tours, Service de Biochimie et de Biologie Moléculaire, Tours, France
| | | |
Collapse
|
29
|
Cao F, Zhou Z, Cai S, Xie W, Jia Z. Hippocampal Long-Term Depression in the Presence of Calcium-Permeable AMPA Receptors. Front Synaptic Neurosci 2018; 10:41. [PMID: 30483111 PMCID: PMC6242858 DOI: 10.3389/fnsyn.2018.00041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/22/2018] [Indexed: 11/28/2022] Open
Abstract
The GluA2 subunit of AMPA glutamate receptors (AMPARs) has been shown to be critical for the expression of NMDA receptor (NMDAR)-dependent long-term depression (LTD). However, in young GluA2 knockout (KO) mice, this form of LTD can still be induced in the hippocampus, suggesting that LTD mechanisms may be modified in the presence of GluA2-lacking, Ca2+ permeable AMPARs. In this study, we examined LTD at the CA1 synapse in GluA2 KO mice by using several well-established inhibitory peptides known to block LTD in wild type (WT) rodents. We showed that while LTD in the KO mice is still blocked by the protein interacting with C kinase 1 (PICK1) peptide pepEVKI, it becomes insensitive to the N-ethylmaleimide-sensitive factor (NSF) peptide pep2m. In addition, the effects of actin and cofilin inhibitory peptides were also altered. These results indicate that in the absence of GluA2, LTD expression mechanisms are different from those in WT animals, suggesting that there are multiple molecular processes enabling LTD expression that are adaptable to physiological and genetic manipulations.
Collapse
Affiliation(s)
- Feng Cao
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Zikai Zhou
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada.,The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,The Collaborative Innovation Center for Brain Science, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Sammy Cai
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,The Collaborative Innovation Center for Brain Science, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Zhengping Jia
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Bie B, Wu J, Foss JF, Naguib M. Amyloid fibrils induce dysfunction of hippocampal glutamatergic silent synapses. Hippocampus 2018; 28:549-556. [PMID: 29704282 PMCID: PMC6133714 DOI: 10.1002/hipo.22955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/14/2018] [Accepted: 04/24/2018] [Indexed: 11/09/2022]
Abstract
Silent glutamatergic synapses lacking functional AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazoleproprionate) receptors exist in several brain regions including the hippocampus. Their involvement in the dysfunction of hippocampal glutamatergic transmission in the setting of Alzheimer's disease (AD) is unknown. This study demonstrated a decrease in the percentage of silent synapses in rats microinjected with amyloid fibrils (Aβ1-40 ) into the hippocampal CA1. Also, pairing low-frequency electric stimuli failed to induce activation of the hippocampal silent synapses in the modeled rats. Immunoblotting studies revealed a decreased expression of GluR1 subunits in the hippocampal CA1 synaptosomal preparation, indicating a potential reduction in the GluR1 subunits anchoring in postsynaptic density in the modeled rats. We also noted a decreased expression of phosphorylated cofilin, which regulates the function of actin cytoskeleton and receptor trafficking, and reduced expression of the scaffolding protein PSD95 in the hippocampal CA1 synaptosome in rats injected with Aβ1-40 . Taken together, this study illustrates dysfunction of hippocampal silent synapse in the rodent model of AD, which might result from the impairments of actin cytoskeleton and postsynaptic scaffolding proteins induced by amyloid fibrils.
Collapse
Affiliation(s)
- Bihua Bie
- Anesthesiology Institute, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave. – NB3-78, Cleveland, OH 44195
| | - Jiang Wu
- Anesthesiology Institute, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave. – NB3-78, Cleveland, OH 44195
| | - Joseph F. Foss
- Anesthesiology Institute, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave. – NB3-78, Cleveland, OH 44195
| | - Mohamed Naguib
- Anesthesiology Institute, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave. – NB3-78, Cleveland, OH 44195
| |
Collapse
|
31
|
Semaphorin 3A as an inhibitive factor for migration of olfactory ensheathing cells through cofilin activation is involved in formation of olfactory nerve layer. Mol Cell Neurosci 2018; 92:27-39. [PMID: 29940213 DOI: 10.1016/j.mcn.2018.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/23/2018] [Accepted: 06/19/2018] [Indexed: 01/27/2023] Open
Abstract
Olfactory ensheathing cells (OECs) migrate from olfactory epithelium towards olfactory bulb (OB), contributing to formation of the presumptive olfactory nerve layer during development. However, it remains unclear that molecular mechanism of regulation of OEC migration in OB. In the present study, we found that OECs highly expressed the receptors of semaphorin 3A (Sema3A) in vitro and in vivo, whereas Sema3A displayed a gradient expression pattern with higher in inner layer of OB and lower in outer layer of OB. Furthermore, the collapse assays, Boyden chamber migration assays and single-cell migration assays showed that Sema3A induced the collapse of leading front of OECs and inhibited OEC migration. Thirdly, the leading front of OECs exhibited adaptation in a protein synthesis-independent manner, and endocytosis-dependent manner during Sema3A-induced OEC migration. Finally, Sema3A-induced collapse of leading front was required the decrease of focal adhesion and a retrograde F-actin flow in a cofilin activation-dependent manner. Taken together, these results demonstrate that Sema3A as an inhibitive migratory factor for OEC migration through cofilin activation is involved in the formation of olfactory nerve layer.
Collapse
|
32
|
Wilson JL, Warburton R, Taylor L, Toksoz D, Hill N, Polgar P. Unraveling endothelin-1 induced hypercontractility of human pulmonary artery smooth muscle cells from patients with pulmonary arterial hypertension. PLoS One 2018; 13:e0195780. [PMID: 29649319 PMCID: PMC5897024 DOI: 10.1371/journal.pone.0195780] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/29/2018] [Indexed: 01/05/2023] Open
Abstract
Contraction of human pulmonary artery smooth muscle cells (HPASMC) isolated from pulmonary arterial hypertensive (PAH) and normal (non-PAH) subject lungs was determined and measured with real-time electrical impedance. Treatment of HPASMC with vasoactive peptides, endothelin-1 (ET-1) and bradykinin (BK) but not angiotensin II, induced a temporal decrease in the electrical impedance profile mirroring constrictive morphological change of the cells which typically was more robust in PAH as opposed to non-PAH cells. Inhibition with LIMKi3 and a cofilin targeted motif mimicking cell permeable peptide (MMCPP) had no effect on ET-1 induced HPASMC contraction indicating a negligible role for these actin regulatory proteins. On the other hand, a MMCPP blocking the activity of caldesmon reduced ET-1 promoted contraction pointing to a regulatory role of this protein and its activation pathway in HPASMC contraction. Inhibition of this MEK/ERK/p90RSK pathway, which is an upstream regulator of caldesmon phosphorylation, reduced ET-1 induced cell contraction. While the regulation of ET-1 induced cell contraction was found to be similar in PAH and non-PAH cells, a key difference was the response to pharmacological inhibitors and to siRNA knockdown of Rho kinases (ROCK1/ROCK2). The PAH cells required much higher concentrations of inhibitors to abrogate ET-1 induced contractions and their contraction was not affected by siRNA against either ROCK1 or ROCK2. Lastly, blocking of L-type and T-type Ca2+ channels had no effect on ET-1 or BK induced contraction. However, inhibiting the activity of the sarcoplasmic reticulum Ca2+ ATPase blunted ET-1 and BK induced HPASMC contraction in both PAH and non-PAH derived HPASMC. In summary, our findings here together with previous communications illustrate similarities and differences in the regulation PAH and non-PAH smooth muscle cell contraction relating to calcium translocation, RhoA/ROCK signaling and the activity of caldesmon. These findings may provide useful tools in achieving the regulation of the vascular hypercontractility taking place in PAH.
Collapse
Affiliation(s)
- Jamie L. Wilson
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| | - Rod Warburton
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Linda Taylor
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Deniz Toksoz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Nicholas Hill
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Peter Polgar
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| |
Collapse
|
33
|
Choi JH, Wang W, Park D, Kim SH, Kim KT, Min KT. IRES-mediated translation of cofilin regulates axonal growth cone extension and turning. EMBO J 2018; 37:embj.201695266. [PMID: 29440227 DOI: 10.15252/embj.201695266] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/19/2017] [Accepted: 01/05/2018] [Indexed: 11/09/2022] Open
Abstract
In neuronal development, dynamic rearrangement of actin promotes axonal growth cone extension, and spatiotemporal translation of local mRNAs in response to guidance cues directs axonal growth cone steering, where cofilin plays a critical role. While regulation of cofilin activity is well studied, regulatory mechanism for cofilin mRNA translation in neurons is unknown. In eukaryotic cells, proteins can be synthesized by cap-dependent or cap-independent mechanism via internal ribosome entry site (IRES)-mediated translation. IRES-mediated translation has been reported in various pathophysiological conditions, but its role in normal physiological environment is poorly understood. Here, we report that 5'UTR of cofilin mRNA contains an IRES element, and cofilin is predominantly translated by IRES-mediated mechanism in neurons. Furthermore, we show that IRES-mediated translation of cofilin is required for both axon extension and axonal growth cone steering. Our results provide new insights into the function of IRES-mediated translation in neuronal development.
Collapse
Affiliation(s)
- Jung-Hyun Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Wei Wang
- Department of Biological Sciences, School of Life Sciences, Ulsan, Korea.,National Creative Research Initiative Center for Proteostasis, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Dongkeun Park
- Department of Biological Sciences, School of Life Sciences, Ulsan, Korea.,National Creative Research Initiative Center for Proteostasis, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Sung-Hoon Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Korea
| | - Kyong-Tai Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea .,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Korea
| | - Kyung-Tai Min
- Department of Biological Sciences, School of Life Sciences, Ulsan, Korea .,National Creative Research Initiative Center for Proteostasis, Ulsan National Institute of Science and Technology, Ulsan, Korea
| |
Collapse
|
34
|
Murakami G, Hojo Y, Kato A, Komatsuzaki Y, Horie S, Soma M, Kim J, Kawato S. Rapid nongenomic modulation by neurosteroids of dendritic spines in the hippocampus: Androgen, oestrogen and corticosteroid. J Neuroendocrinol 2018; 30. [PMID: 29194818 DOI: 10.1111/jne.12561] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/22/2022]
Abstract
Memories are stored in synapses that consist of axon terminals and dendritic spines. Dendritic spines are postsynaptic structures of synapses and are essential for synaptic plasticity and cognition. Therefore, extensive investigations concerning the functions and structures of spines have been performed. Sex steroids and stress steroids have been shown to modulate hippocampal synapses. Although the rapid modulatory action of sex steroids on synapses has been studied in hippocampal neurones over several decades, the essential molecular mechanisms have not been fully understood. Here, a description of kinase-dependent signalling mechanisms is provided that can explain the rapid nongenomic modulation of dendritic spinogenesis in rat and mouse hippocampal slices by the application of sex steroids, including dihydrotestosterone, testosterone, oestradiol and progesterone. We also indicate the role of synaptic (classic) sex steroid receptors that trigger these rapid synaptic modulations. Moreover, we describe rapid nongenomic spine modulation by applying corticosterone, which is an acute stress model of the hippocampus. The explanations for the results obtained are mainly based on the optical imaging of dendritic spines. Comparisons are also performed with results obtained from other types of imaging, including electron microscopic imaging. Relationships between spine modulation and modulation of cognition are discussed. We recognise that most of rapid effects of exogenously applied oestrogen and androgen were observed in steroid-depleted conditions, including acute slices of the hippocampus, castrated male animals and ovariectomised female animals. Therefore, the previously observed effects can be considered as a type of recovery event, which may be essentially similar to hormone replacement therapy under hormone-decreased conditions. On the other hand, in gonadally intact young animals with high levels of endogenous sex hormones, further supplementation of sex hormones might not be effective, whereas the infusion of blockers for steroid receptors or kinases may be effective, with respect to suppressing sex hormone functions, thus providing useful information regarding molecular mechanisms.
Collapse
Affiliation(s)
- G Murakami
- Department of Liberal Arts, Faculty of Medicine, Saitama Medical University, Iruma, Saitama, Japan
| | - Y Hojo
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Iruma, Saitama, Japan
| | - A Kato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Meguro, Tokyo, Japan
| | - Y Komatsuzaki
- Department of Physics, College of Science and Technology, Nihon University, Chiyoda, Tokyo, Japan
| | - S Horie
- Department of Urology, Graduate School of Medicine, Juntendo University, Hongo, Tokyo, Japan
| | - M Soma
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Itabashi, Tokyo, Japan
| | - J Kim
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Itabashi, Tokyo, Japan
| | - S Kawato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Meguro, Tokyo, Japan
- Department of Urology, Graduate School of Medicine, Juntendo University, Hongo, Tokyo, Japan
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Itabashi, Tokyo, Japan
| |
Collapse
|
35
|
Deng Y, Wei J, Cheng J, Zhong P, Xiong Z, Liu A, Lin L, Chen S, Yan Z. Partial Amelioration of Synaptic and Cognitive Deficits by Inhibiting Cofilin Dephosphorylation in an Animal Model of Alzheimer's Disease. J Alzheimers Dis 2018; 53:1419-32. [PMID: 27372643 DOI: 10.3233/jad-160167] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The loss of synaptic structure and function has been linked to the cognitive impairment of Alzheimer's disease (AD). Dysregulation of the actin cytoskeleton, which plays a key role in regulating the integrity of synapses and the transport of synaptic proteins, has been suggested to contribute to the pathology of AD. In this study, we found that glutamate receptor surface expression and synaptic function in frontal cortical neurons were significant diminished in a familial AD (FAD) model, which was correlated with the reduction of phosphorylated cofilin, a key protein regulating the dynamics of actin filaments. Injecting a cofilin dephosphorylation inhibitory peptide to FAD mice led to the partial rescue of the surface expression of AMPA and NMDA receptor subunits, as well as the partial restoration of AMPAR- and NMDAR-mediated synaptic currents. Moreover, the impaired working memory and novel object recognition memory in FAD mice were partially ameliorated by injections of the cofilin dephosphorylation inhibitory peptide. These results suggest that targeting the cofilin-actin signaling holds promise to mitigate the physiological and behavioral abnormality in AD.
Collapse
Affiliation(s)
- Yulei Deng
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wei
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, USA.,VA Western New York Healthcare System, Buffalo, NY, USA
| | - Jia Cheng
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Ping Zhong
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, USA.,VA Western New York Healthcare System, Buffalo, NY, USA
| | - Zhe Xiong
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Aiyi Liu
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lin Lin
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Shengdi Chen
- Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Yan
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, USA.,VA Western New York Healthcare System, Buffalo, NY, USA
| |
Collapse
|
36
|
Schlau M, Terheyden-Keighley D, Theis V, Mannherz HG, Theiss C. VEGF Triggers the Activation of Cofilin and the Arp2/3 Complex within the Growth Cone. Int J Mol Sci 2018; 19:ijms19020384. [PMID: 29382077 PMCID: PMC5855606 DOI: 10.3390/ijms19020384] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/16/2018] [Accepted: 01/24/2018] [Indexed: 01/05/2023] Open
Abstract
A crucial neuronal structure for the development and regeneration of neuronal networks is the axonal growth cone. Affected by different guidance cues, it grows in a predetermined direction to reach its final destination. One of those cues is the vascular endothelial growth factor (VEGF), which was identified as a positive effector for growth cone movement. These positive effects are mainly mediated by a reorganization of the actin network. This study shows that VEGF triggers a tight colocalization of cofilin and the Arp2/3 complex to the actin cytoskeleton within chicken dorsal root ganglia (DRG). Live cell imaging after microinjection of GFP (green fluorescent protein)-cofilin and RFP (red fluorescent protein)-LifeAct revealed that both labeled proteins rapidly redistributed within growth cones, and showed a congruent distribution pattern after VEGF supplementation. Disruption of signaling upstream of cofilin via blocking LIM-kinase (LIMK) activity resulted in growth cones displaying regressive growth behavior. Microinjection of GFP-p16b (a subunit of the Arp2/3 complex) and RFP-LifeAct revealed that both proteins redistributed into lamellipodia of the growth cone within minutes after VEGF stimulation. Disruption of the signaling to the Arp2/3 complex in the presence of VEGF by inhibition of N-WASP (neuronal Wiskott–Aldrich–Scott protein) caused retraction of growth cones. Hence, cofilin and the Arp2/3 complex appear to be downstream effector proteins of VEGF signaling to the actin cytoskeleton of DRG growth cones. Our data suggest that VEGF simultaneously affects different pathways for signaling to the actin cytoskeleton, since activation of cofilin occurs via inhibition of LIMK, whereas activation of Arp2/3 is achieved by stimulation of N-WASP.
Collapse
Affiliation(s)
- Matthias Schlau
- Institute of Anatomy, Department of Cytology, Ruhr-University Bochum, Universitätsstraße 150, 44780 Bochum, Germany.
| | - Daniel Terheyden-Keighley
- Institute of Anatomy, Department of Cytology, Ruhr-University Bochum, Universitätsstraße 150, 44780 Bochum, Germany.
| | - Verena Theis
- Institute of Anatomy, Department of Cytology, Ruhr-University Bochum, Universitätsstraße 150, 44780 Bochum, Germany.
| | - Hans Georg Mannherz
- Research Group Molecular Cardiology, University Hospital Bergmannsheil and St. Josef Hospital, c/o Clinical Pharmacology, Ruhr-University, 44780 Bochum, Germany.
| | - Carsten Theiss
- Institute of Anatomy, Department of Cytology, Ruhr-University Bochum, Universitätsstraße 150, 44780 Bochum, Germany.
| |
Collapse
|
37
|
Hojo Y, Kawato S. Neurosteroids in Adult Hippocampus of Male and Female Rodents: Biosynthesis and Actions of Sex Steroids. Front Endocrinol (Lausanne) 2018; 9:183. [PMID: 29740398 PMCID: PMC5925962 DOI: 10.3389/fendo.2018.00183] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
The brain is not only the target of steroid hormones but also is able to locally synthesize steroids de novo. Evidence of the local production of steroids in the brain has been accumulating in various vertebrates, including teleost fish, amphibia, birds, rodents, non-human primates, and humans. In this review, we mainly focus on the local production of sex steroids in the hippocampal neurons of adult rodents (rats and mice), a center for learning and memory. From the data of the hippocampus of adult male rats, hippocampal principal neurons [pyramidal cells in CA1-CA3 and granule cells in dentate gyrus (DG)] have a complete system for biosynthesis of sex steroids. Liquid chromatography with tandem-mass-spectrometry (LC-MS/MS) enabled us to accurately determine the levels of hippocampal sex steroids including 17β-estradiol (17β-E2), testosterone (T), and dihydrotestosterone (DHT), which are much higher than those in blood. Next, we review the steroid synthesis in the hippocampus of female rats, since previous knowledge had been biased toward the data from males. Recently, we clarified that the levels of hippocampal steroids fluctuate in adult female rats across the estrous cycle. Accurate determination of hippocampal steroids at each stage of the estrous cycle is of importance for providing the account for the fluctuation of female hippocampal functions, including spine density, long-term potentiation (LTP) and long-term depression (LTD), and learning and memory. These functional fluctuations in female had been attributed to the level of circulation-derived steroids. LC-MS/MS analysis revealed that the dendritic spine density in CA1 of adult female hippocampus correlates with the levels of hippocampal progesterone and 17β-E2. Finally, we introduce the direct evidence of the role of hippocampus-synthesized steroids in hippocampal function including neurogenesis, LTP, and memory consolidation. Mild exercise (2 week of treadmill running) elevated synthesis of DHT in the hippocampus, but not in the testis, of male rats, resulting in enhancement of neurogenesis in DG. Concerning synaptic plasticity, hippocampus-synthesized E2 is required for LTP induction, whereas hippocampus-synthesized DHT is required for LTD induction. Furthermore, hippocampus-synthesized E2 is involved in memory consolidation tested by object recognition and object placement tasks, both of which are hippocampus-dependent.
Collapse
Affiliation(s)
- Yasushi Hojo
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
- *Correspondence: Yasushi Hojo,
| | - Suguru Kawato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| |
Collapse
|
38
|
Spurlin JW, Nelson CM. Building branched tissue structures: from single cell guidance to coordinated construction. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2015.0527. [PMID: 28348257 DOI: 10.1098/rstb.2015.0527] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2016] [Indexed: 12/15/2022] Open
Abstract
Branched networks are ubiquitous throughout nature, particularly found in tissues that require large surface area within a restricted volume. Many tissues with a branched architecture, such as the vasculature, kidney, mammary gland, lung and nervous system, function to exchange fluids, gases and information throughout the body of an organism. The generation of branched tissues requires regulation of branch site specification, initiation and elongation. Branching events often require the coordination of many cells to build a tissue network for material exchange. Recent evidence has emerged suggesting that cell cooperativity scales with the number of cells actively contributing to branching events. Here, we compare mechanisms that regulate branching, focusing on how cell cohorts behave in a coordinated manner to build branched tissues.This article is part of the themed issue 'Systems morphodynamics: understanding the development of tissue hardware'.
Collapse
Affiliation(s)
- James W Spurlin
- Departments of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Departments of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA .,Molecular Biology, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA
| |
Collapse
|
39
|
Kung F, Wang W, Tran TS, Townes-Anderson E. Sema3A Reduces Sprouting of Adult Rod Photoreceptors In Vitro. Invest Ophthalmol Vis Sci 2017; 58:4318–4331. [PMID: 28806446 PMCID: PMC5555408 DOI: 10.1167/iovs.16-21075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Purpose Rod photoreceptor terminals respond to retinal injury with retraction and sprouting. Since the guidance cue Semaphorin3A (Sema3A) is observed in the retina after injury, we asked whether Sema3A contributes to structural plasticity in rod photoreceptors. Methods We used Western blots and alkaline phosphatase (AP)-tagged neuropilin-1 (NPN-1) to detect the expression of Sema3A in an organotypic model of porcine retinal detachment. We then examined Sema3A binding to cultured salamander rod photoreceptors using AP-tagged Sema3A. For functional analysis, we used a microspritzer to apply a gradient of Sema3A-Fc to isolated salamander rod photoreceptors over 24 hours. Results Sema3A protein was biochemically detected in porcine retinal explants in the retina 7, 24, and 72 hours after detachment. In sections, NPN-1 receptor was bound to the inner and outer retina. For isolated rod photoreceptors, Sema3A localized to synaptic terminals and to neuritic processes after 1 week in vitro. In microspritzed rod photoreceptors, process initiation occurred away from high concentrations of Sema3A. Sema3A significantly decreased the number of processes formed by rod photoreceptors although the average length of processes was not affected. The cellular orientation of rod photoreceptors relative to the microspritzer also significantly changed over time; this effect was reduced with the Sema3A inhibitor, xanthofulvin. Conclusion Sema3A is expressed in the retina after detachment, binds to rod photoreceptors, affects cell orientation, and reduces photoreceptor process initiation in vitro. Our results suggest that Sema3A contributes to axonal retraction in retinal injury, whereas rod neuritic sprouting and regenerative synaptogenesis may require a reduction in semaphorin signaling.
Collapse
Affiliation(s)
- Frank Kung
- Joint Program in Biomedical Engineering, Rutgers University, Graduate School of Biomedical Sciences, New Jersey Institute of Technology, Newark, United States
| | - Weiwei Wang
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University, New Jersey Medical School, Newark, New Jersey, United States
| | - Tracy S Tran
- Department of Biological Sciences, Rutgers University, Newark College of Arts and Sciences, Newark, New Jersey, United States
| | - Ellen Townes-Anderson
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University, New Jersey Medical School, Newark, New Jersey, United States
| |
Collapse
|
40
|
Shaw AE, Bamburg JR. Peptide regulation of cofilin activity in the CNS: A novel therapeutic approach for treatment of multiple neurological disorders. Pharmacol Ther 2017; 175:17-27. [PMID: 28232023 PMCID: PMC5466456 DOI: 10.1016/j.pharmthera.2017.02.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cofilin is a ubiquitous protein which cooperates with many other actin-binding proteins in regulating actin dynamics. Cofilin has essential functions in nervous system development including neuritogenesis, neurite elongation, growth cone pathfinding, dendritic spine formation, and the regulation of neurotransmission and spine function, components of synaptic plasticity essential for learning and memory. Cofilin's phosphoregulation is a downstream target of many transmembrane signaling processes, and its misregulation in neurons has been linked in rodent models to many different neurodegenerative and neurological disorders including Alzheimer disease (AD), aggression due to neonatal isolation, autism, manic/bipolar disorder, and sleep deprivation. Cognitive and behavioral deficits of these rodent models have been largely abrogated by modulation of cofilin activity using viral-mediated, genetic, and/or small molecule or peptide therapeutic approaches. Neuropathic pain in rats from sciatic nerve compression has also been reduced by modulating the cofilin pathway within neurons of the dorsal root ganglia. Neuroinflammation, which occurs following cerebral ischemia/reperfusion, but which also accompanies many other neurodegenerative syndromes, is markedly reduced by peptides targeting specific chemokine receptors, which also modulate cofilin activity. Thus, peptide therapeutics offer potential for cost-effective treatment of a wide variety of neurological disorders. Here we discuss some recent results from rodent models using therapeutic peptides with a surprising ability to cross the rodent blood brain barrier and alter cofilin activity in brain. We also offer suggestions as to how neuronal-specific cofilin regulation might be achieved.
Collapse
Affiliation(s)
- Alisa E Shaw
- Department of Biochemistry and Molecular Biology, Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523-1870, United States
| | - James R Bamburg
- Department of Biochemistry and Molecular Biology, Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523-1870, United States.
| |
Collapse
|
41
|
Quantitative Phosphoproteomics Reveals a Role for Collapsin Response Mediator Protein 2 in PDGF-Induced Cell Migration. Sci Rep 2017. [PMID: 28638064 PMCID: PMC5479788 DOI: 10.1038/s41598-017-04015-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The Platelet Derived Growth Factor (PDGF) family of ligands have well established functions in the induction of cell proliferation and migration during development, tissue homeostasis and interactions between tumours and stroma. However, the mechanisms by which these actions are executed are incompletely understood. Here we report a differential phosphoproteomics study, using a SILAC approach, of PDGF-stimulated mouse embryonic fibroblasts (MEFs). 116 phospho-sites were identified as up-regulated and 45 down-regulated in response to PDGF stimulation. These encompass proteins involved in cell adhesion, cytoskeleton regulation and vesicle-mediated transport, significantly expanding the range of proteins implicated in PDGF signalling pathways. Included in the down-regulated class was the microtubule bundling protein Collapsin Response Mediator Protein 2 (CRMP2). In response to stimulation with PDGF, CRMP2 was dephosphorylated on Thr514, an event known to increase CRMP2 activity. This was reversed in the presence of micromolar concentrations of the protein phosphatase inhibitor okadaic acid, implicating PDGF-induced activation of protein phosphatase 1 (PP1) in CRMP2 regulation. Depletion of CRMP2 resulted in impairment of PDGF-mediated cell migration in an in vitro wound healing assay. These results show that CRMP2 is required for PDGF-directed cell migration in vitro.
Collapse
|
42
|
Vogel Ciernia A, Kramár EA, Matheos DP, Havekes R, Hemstedt TJ, Magnan CN, Sakata K, Tran A, Azzawi S, Lopez A, Dang R, Wang W, Trieu B, Tong J, Barrett RM, Post RJ, Baldi P, Abel T, Lynch G, Wood MA. Mutation of neuron-specific chromatin remodeling subunit BAF53b: rescue of plasticity and memory by manipulating actin remodeling. Learn Mem 2017; 24:199-209. [PMID: 28416631 PMCID: PMC5397687 DOI: 10.1101/lm.044602.116] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
Recent human exome-sequencing studies have implicated polymorphic Brg1-associated factor (BAF) complexes (mammalian SWI/SNF chromatin remodeling complexes) in several intellectual disabilities and cognitive disorders, including autism. However, it remains unclear how mutations in BAF complexes result in impaired cognitive function. Post-mitotic neurons express a neuron-specific assembly, nBAF, characterized by the neuron-specific subunit BAF53b. Subdomain 2 of BAF53b is essential for the differentiation of neuronal precursor cells into neurons. We generated transgenic mice lacking subdomain 2 of Baf53b (BAF53bΔSB2). Long-term synaptic potentiation (LTP) and long-term memory, both of which are associated with phosphorylation of the actin severing protein cofilin, were assessed in these animals. A phosphorylation mimic of cofilin was stereotaxically delivered into the hippocampus of BAF53bΔSB2 mice in an effort to rescue LTP and memory. BAF53bΔSB2 mutant mice show impairments in phosphorylation of synaptic cofilin, LTP, and memory. Both the synaptic plasticity and memory deficits are rescued by overexpression of a phosphorylation mimetic of cofilin. Baseline physiology and behavior were not affected by the mutation or the experimental treatment. This study suggests a potential link between nBAF function, actin cytoskeletal remodeling at the dendritic spine, and memory formation. This work shows that a targeted manipulation of synaptic function can rescue adult plasticity and memory deficits caused by manipulations of nBAF, and thereby provides potential novel avenues for therapeutic development for multiple intellectual disability disorders.
Collapse
Affiliation(s)
- Annie Vogel Ciernia
- Department of Medical Microbiology and Immunology, University of California, Davis, California 95656, USA
| | - Enikö A Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Dina P Matheos
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Robbert Havekes
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen 9712, The Netherlands
| | - Thekla J Hemstedt
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Christophe N Magnan
- Department of Computer Science, University of California, Irvine, California 92697, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697, USA
| | - Keith Sakata
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Ashley Tran
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Soraya Azzawi
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Alberto Lopez
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Richard Dang
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Weisheng Wang
- Department of Computer Science, University of California, Irvine, California 92697, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697, USA
| | - Brian Trieu
- Department of Anatomy and Neurobiology, University of California, Irvine, California 92697, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697, USA
| | - Joyce Tong
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Ruth M Barrett
- Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Rebecca J Post
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
| | - Pierre Baldi
- Department of Computer Science, University of California, Irvine, California 92697, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697, USA
| | - Ted Abel
- Departments of Molecular Physiology and Biophysics, Psychiatry, and Biochemistry, Iowa Neuroscience Institute, Iowa City, Iowa 50309, USA
| | - Gary Lynch
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697, USA
- Center for the Neurobiology of Learning and Memory, Irvine, California, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697, USA
| |
Collapse
|
43
|
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: 81] [Impact Index Per Article: 10.1] [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
|
44
|
Yamane M, Yamashita N, Hida T, Kamiya Y, Nakamura F, Kolattukudy P, Goshima Y. A functional coupling between CRMP1 and Na v1.7 for retrograde propagation of Semaphorin3A signaling. J Cell Sci 2017; 130:1393-1403. [PMID: 28254884 DOI: 10.1242/jcs.199737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/22/2017] [Indexed: 12/19/2022] Open
Abstract
Semaphorin3A (Sema3A) is a secreted type of axon guidance molecule that regulates axon wiring through complexes of neuropilin-1 (NRP1) with PlexinA protein receptors. Sema3A regulates the dendritic branching through tetrodotoxin (TTX)-sensitive retrograde axonal transport of PlexA proteins and tropomyosin-related kinase A (TrkA) complex. We here demonstrate that Nav1.7 (encoded by SCN9A), a TTX-sensitive Na+ channel, by coupling with collapsin response mediator protein 1 (CRMP1), mediates the Sema3A-induced retrograde transport. In mouse dorsal root ganglion (DRG) neurons, Sema3A increased co-localization of PlexA4 and TrkA in the growth cones and axons. TTX treatment and RNAi knockdown of Nav1.7 sustained Sema3A-induced colocalized signals of PlexA4 and TrkA in growth cones and suppressed the subsequent localization of PlexA4 and TrkA in distal axons. A similar localization phenotype was observed in crmp1-/- DRG neurons. Sema3A induced colocalization of CRMP1 and Nav1.7 in the growth cones. The half maximal voltage was increased in crmp1-/- neurons when compared to that in wild type. In HEK293 cells, introduction of CRMP1 lowered the threshold of co-expressed exogenous Nav1.7. These results suggest that Nav1.7, by coupling with CRMP1, mediates the axonal retrograde signaling of Sema3A.
Collapse
Affiliation(s)
- Masayuki Yamane
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan .,Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tomonobu Hida
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan.,RIKEN Brain Science Institute, Saitama 351-0198, Japan
| | - Yoshinori Kamiya
- Department of Anesthesiology, Uonuma Institute of Community Medicine, Niigata University Medical and Dental Hospital, 4132 Urasa, Minami-uonuma, Niigata 949-7302, Japan
| | - Fumio Nakamura
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Pappachan Kolattukudy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
45
|
Valek L, Häussler A, Dröse S, Eaton P, Schröder K, Tegeder I. Redox-guided axonal regrowth requires cyclic GMP dependent protein kinase 1: Implication for neuropathic pain. Redox Biol 2016; 11:176-191. [PMID: 27978504 PMCID: PMC5156608 DOI: 10.1016/j.redox.2016.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/03/2016] [Accepted: 12/02/2016] [Indexed: 01/27/2023] Open
Abstract
Cyclic GMP-dependent protein kinase 1 (PKG1) mediates presynaptic nociceptive long-term potentiation (LTP) in the spinal cord and contributes to inflammatory pain in rodents but the present study revealed opposite effects in the context of neuropathic pain. We used a set of loss-of-function models for in vivo and in vitro studies to address this controversy: peripheral neuron specific deletion (SNS-PKG1-/-), inducible deletion in subsets of neurons (SLICK-PKG1-/-) and redox-dead PKG1 mutants. In contrast to inflammatory pain, SNS-PKG1-/- mice developed stronger neuropathic hyperalgesia associated with an impairment of nerve regeneration, suggesting specific repair functions of PKG1. Although PKG1 accumulated at the site of injury, its activity was lost in the proximal nerve due to a reduction of oxidation-dependent dimerization, which was a consequence of mitochondrial damage in injured axons. In vitro, PKG1 deficiency or its redox-insensitivity resulted in enhanced outgrowth and reduction of growth cone collapse in response to redox signals, which presented as oxidative hotspots in growing cones. At the molecular level, PKG1 deficiency caused a depletion of phosphorylated cofilin, which is essential for growth cone collapse and guidance. Hence, redox-mediated guidance required PKG1 and consequently, its deficiency in vivo resulted in defective repair and enhanced neuropathic pain after nerve injury. PKG1-dependent repair functions will outweigh its signaling functions in spinal nociceptive LTP, so that inhibition of PKG1 is no option for neuropathic pain. Axonal injury leads mitochondrial damage. The loss of signaling ROS is associated with a reduction of redox-dependent autoactivation of PKG1. Loss of PKG1 impairs peripheral nerve regeneration and aggravates neuropathic pain in mice. Oxidative hot spots are generated in spiky growth cones and trigger growth cone collapse via PKG1. Malfunctioning of this redox-PKG1 guided growth cone collapse leads to aberrant outgrowth.
Collapse
Affiliation(s)
- Lucie Valek
- Depts. of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Annett Häussler
- Depts. of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Stefan Dröse
- Depts. of Anaesthesiology, Goethe-University Hospital, Frankfurt, Germany
| | - Philipp Eaton
- King's College of London, Cardiovascular Division, The Rayne Institute, St. Thomas' Hospital, London, United Kingdom
| | - Katrin Schröder
- Depts. of Cardiovascular Physiology, Goethe-University Hospital, Frankfurt, Germany
| | - Irmgard Tegeder
- Depts. of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany.
| |
Collapse
|
46
|
Cao F, Zhou Z, Pan X, Leung C, Xie W, Collingridge G, Jia Z. Developmental regulation of hippocampal long-term depression by cofilin-mediated actin reorganization. Neuropharmacology 2016; 112:66-75. [PMID: 27543417 DOI: 10.1016/j.neuropharm.2016.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/15/2016] [Accepted: 08/15/2016] [Indexed: 01/16/2023]
Abstract
Long lasting synaptic plasticity involves both functional and morphological changes, but how these processes are molecularly linked to achieve coordinated plasticity remains poorly understood. Cofilin is a common target of multiple signaling pathways at the synapse and is required for both functional and spine plasticity, but how it is regulated is unclear. In this study, we investigate whether the involvement of cofilin in plasticity is developmentally regulated by examining the role of cofilin in hippocampal long-term depression (LTD) in both young (2 weeks) and mature (2 months) mice. We show that both total protein level of cofilin and its activation undergo significant changes as the brain matures, so that although the amount of cofilin decreases significantly in mature mice, its regulation by protein phosphorylation becomes increasingly important. Consistent with these biochemical data, we show that cofilin-mediated actin reorganization is essential for LTD in mature, but not in young mice. In contrast to cofilin, the GluA2 interactions with NSF and PICK1 appear to be required in both young and mature mice, indicating that AMPAR internalization is a common key mechanism for LTD expression regardless of the developmental stages. These results establish the temporal specificity of cofilin in LTD regulation and suggest that cofilin-mediated actin reorganization may serve as a key mechanism underlying developmental regulation of synaptic and spine plasticity. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.
Collapse
Affiliation(s)
- Feng Cao
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., Toronto, Ontario M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Canada; The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Zikai Zhou
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., Toronto, Ontario M5G 1X8, Canada; The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China; Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, Nanjing 210096, China
| | - Xingxiu Pan
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Celeste Leung
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., Toronto, Ontario M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Canada
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China; Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, Nanjing 210096, China
| | - Graham Collingridge
- Department of Physiology, Faculty of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Canada; Centre for Synaptic Plasticity, School of Physiology & Pharmacology, University of Bristol, UK
| | - Zhengping Jia
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., Toronto, Ontario M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Canada.
| |
Collapse
|
47
|
Liu A, Zhou Z, Dang R, Zhu Y, Qi J, He G, Leung C, Pak D, Jia Z, Xie W. Neuroligin 1 regulates spines and synaptic plasticity via LIMK1/cofilin-mediated actin reorganization. J Cell Biol 2016; 212:449-63. [PMID: 26880202 PMCID: PMC4754719 DOI: 10.1083/jcb.201509023] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The C-terminal domain of NLG1 is sufficient to enhance spine and synapse number and to modulate synaptic plasticity, and it exerts these effects via its interaction with SPAR and the subsequent activation of LIMK1/cofilin-mediated actin reorganization. Neuroligin (NLG) 1 is important for synapse development and function, but the underlying mechanisms remain unclear. It is known that at least some aspects of NLG1 function are independent of the presynaptic neurexin, suggesting that the C-terminal domain (CTD) of NLG1 may be sufficient for synaptic regulation. In addition, NLG1 is subjected to activity-dependent proteolytic cleavage, generating a cytosolic CTD fragment, but the significance of this process remains unknown. In this study, we show that the CTD of NLG1 is sufficient to (a) enhance spine and synapse number, (b) modulate synaptic plasticity, and (c) exert these effects via its interaction with spine-associated Rap guanosine triphosphatase–activating protein and subsequent activation of LIM-domain protein kinase 1/cofilin–mediated actin reorganization. Our results provide a novel postsynaptic mechanism by which NLG1 regulates synapse development and function.
Collapse
Affiliation(s)
- An Liu
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Zikai Zhou
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, Nanjing 210096, China
| | - Rui Dang
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Yuehua Zhu
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Junxia Qi
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Guiqin He
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Celeste Leung
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Daniel Pak
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20007
| | - Zhengping Jia
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, Nanjing 210096, China
| |
Collapse
|
48
|
Grintsevich EE, Yesilyurt HG, Rich SK, Hung RJ, Terman JR, Reisler E. F-actin dismantling through a redox-driven synergy between Mical and cofilin. Nat Cell Biol 2016; 18:876-85. [PMID: 27454820 PMCID: PMC4966907 DOI: 10.1038/ncb3390] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/21/2016] [Indexed: 02/06/2023]
Abstract
Numerous cellular functions depend on actin filament (F-actin) disassembly. The
best-characterized disassembly proteins, the ADF/cofilins/twinstar, sever filaments and
recycle monomers to promote actin assembly. Cofilin is also a relatively weak actin
disassembler, posing questions about mechanisms of cellular F-actin destabilization. Here
we uncover a key link to targeted F-actin disassembly by finding that F-actin is
efficiently dismantled through a post-translational-mediated synergism between cofilin and
the actin-oxidizing enzyme Mical. We find that Mical-mediated oxidation of actin improves
cofilin binding to filaments, where their combined effect dramatically accelerates F-actin
disassembly compared to either effector alone. This synergism is also necessary and
sufficient for F-actin disassembly in vivo, magnifying the effects of
both Mical and cofilin on cellular remodeling, axon guidance, and Semaphorin/Plexin
repulsion. Mical and cofilin, therefore, form a Redox-dependent synergistic pair that
promotes F-actin instability by rapidly dismantling F-actin and generating
post-translationally modified actin that has altered assembly properties.
Collapse
Affiliation(s)
- Elena E Grintsevich
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, California 90095, USA
| | - Hunkar Gizem Yesilyurt
- Departments of Neuroscience and Pharmacology and Neuroscience Graduate Program, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Shannon K Rich
- Departments of Neuroscience and Pharmacology and Neuroscience Graduate Program, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ruei-Jiun Hung
- Departments of Neuroscience and Pharmacology and Neuroscience Graduate Program, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Jonathan R Terman
- Departments of Neuroscience and Pharmacology and Neuroscience Graduate Program, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, California 90095, USA.,Molecular Biology Institute, University of California-Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
49
|
Goshima Y, Yamashita N, Nakamura F, Sasaki Y. Regulation of dendritic development by semaphorin 3A through novel intracellular remote signaling. Cell Adh Migr 2016; 10:627-640. [PMID: 27392015 DOI: 10.1080/19336918.2016.1210758] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Numerous cell adhesion molecules, extracellular matrix proteins and axon guidance molecules participate in neuronal network formation through local effects at axo-dendritic, axo-axonic or dendro-dendritic contact sites. In contrast, neurotrophins and their receptors play crucial roles in neural wiring by sending retrograde signals to remote cell bodies. Semaphorin 3A (Sema3A), a prototype of secreted type 3 semaphorins, is implicated in axon repulsion, dendritic branching and synapse formation via binding protein neuropilin-1 (NRP1) and the signal transducing protein PlexinAs (PlexAs) complex. This review focuses on Sema3A retrograde signaling that regulates dendritic localization of AMPA-type glutamate receptor GluA2 and dendritic patterning. This signaling is elicited by activation of NRP1 in growth cones and is propagated to cell bodies by dynein-dependent retrograde axonal transport of PlexAs. It also requires interaction between PlexAs and a high-affinity receptor for nerve growth factor, toropomyosin receptor kinase A. We propose a control mechanism by which retrograde Sema3A signaling regulates the glutamate receptor localization through trafficking of cis-interacting PlexAs with GluA2 along dendrites; this remote signaling may be an alternative mechanism to local adhesive contacts for neural network formation.
Collapse
Affiliation(s)
- Yoshio Goshima
- a Department of Molecular Pharmacology and Neurobiology , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Naoya Yamashita
- a Department of Molecular Pharmacology and Neurobiology , Yokohama City University Graduate School of Medicine , Yokohama , Japan.,c Department of Biology , Johns Hopkins University , Baltimore , MD , USA
| | - Fumio Nakamura
- a Department of Molecular Pharmacology and Neurobiology , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yukio Sasaki
- b Functional Structural, Biology Laboratory, Department of Medical Life Science , Yokohama City University Graduate School of Medical Life Science , Suehirocho, Tsurumi-ku, Yokohama , Japan
| |
Collapse
|
50
|
Wang W, Rai A, Hur EM, Smilansky Z, Chang KT, Min KT. DSCR1 is required for both axonal growth cone extension and steering. J Cell Biol 2016; 213:451-62. [PMID: 27185837 PMCID: PMC4878092 DOI: 10.1083/jcb.201510107] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/22/2016] [Indexed: 11/26/2022] Open
Abstract
Wang et al. identify that DSCR1, a gene on chromosome 21 that is associated with Down syndrome, controls both the rate and direction of axon growth in response to extrinsic cues by regulating cytoskeletal dynamics and local protein synthesis in the growth cone. Local information processing in the growth cone is essential for correct wiring of the nervous system. As an axon navigates through the developing nervous system, the growth cone responds to extrinsic guidance cues by coordinating axon outgrowth with growth cone steering. It has become increasingly clear that axon extension requires proper actin polymerization dynamics, whereas growth cone steering involves local protein synthesis. However, molecular components integrating these two processes have not been identified. Here, we show that Down syndrome critical region 1 protein (DSCR1) controls axon outgrowth by modulating growth cone actin dynamics through regulation of cofilin activity (phospho/dephospho-cofilin). Additionally, DSCR1 mediates brain-derived neurotrophic factor–induced local protein synthesis and growth cone turning. Our study identifies DSCR1 as a key protein that couples axon growth and pathfinding by dually regulating actin dynamics and local protein synthesis.
Collapse
Affiliation(s)
- Wei Wang
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Korea
| | - Asit Rai
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Korea
| | - Eun-Mi Hur
- Brain Science Institute-Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, Korea Department of Neuroscience, University of Science and Technology, Daejeon 34113, Korea
| | | | - Karen T Chang
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90089 Department of Cell and Neurobiology, University of Southern California, Los Angeles, CA 90089
| | - Kyung-Tai Min
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Korea
| |
Collapse
|