1
|
Majumder T, Khot B, Suriyaarachchi H, Nathan A, Liu G. MYC regulation of the miR-92-Robo1 axis in Slit-mediated commissural axon guidance. Mol Biol Cell 2025; 36:ar50. [PMID: 40020181 PMCID: PMC12005101 DOI: 10.1091/mbc.e24-12-0534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
In the developing spinal cord, translational repression of Robo1 expression by microRNA-92 (miR-92) in precrossing commissural axons (CAs) inhibits Slit/Robo1-mediated repulsion facilitating commissural axon projection and midline crossing; however, the regulatory mechanisms governing miR-92 expression in the developing commissural neurons are currently lacking. Here, we propose that the transcription factor MYC regulates miR-92 expression in the developing spinal cord (of either sex) to control Robo1 levels in precrossing CAs, modulating Slit/Robo1-mediated repulsion and midline crossing. MYC, miR-92, and Robo1 are differentially expressed in the developing chicken spinal cord. MYC binds to the promoter region upstream of the gga-miR-92 gene in vitro. MYC knockdown dramatically decreases miR-92 expression and increases chicken Robo1 (cRobo1) levels. In contrast, overexpression of MYC significantly induces miR-92 expression and reduces cRobo1 levels. MYC knockdown or overexpression results in significant inhibition or induction of miR-92 activity in the developing chicken spinal cord, respectively. Disruption of the MYC-dependent regulation of the miR-92-cRobo1 axis affects Slit2-mediated CA growth cone collapse in vitro and impairs CA projection and midline crossing in vivo. These results elucidate the role of the MYC-miR-92-cRobo1 axis in Slit2/Robo1-mediated CA repulsion and midline crossing.
Collapse
Affiliation(s)
- Tanushree Majumder
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Bhakti Khot
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | | | - Anagaa Nathan
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| |
Collapse
|
2
|
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
|
3
|
Nedaei K, Hesaraki M, Mazloomzadeh S, Totonchi M, Biglari AR. Lentiviral Mediated Expression of Soluble Neuropilin 1 Inhibits Semaphorin 3A-mediated Collapse Activity in Vitro. Basic Clin Neurosci 2021; 12:223-232. [PMID: 34925719 PMCID: PMC8672662 DOI: 10.32598/bcn.12.2.1678.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/14/2020] [Accepted: 08/02/2020] [Indexed: 11/20/2022] Open
Abstract
Introduction Semaphorin 3A (Sema 3A) is a secreted protein, which plays an integral part in developing the nervous system. It has collapse activity on the growth cone of dorsal root ganglia. After the development of the nervous system, Sema 3A expression decreases. Neuropilin 1 is a membrane receptor of Sema 3A. When semaphorin binds to neuropilin 1, the recruitment of oligodendrocyte precursor cells to the demyelinated site decreases. In Multiple Sclerosis (MS), Sema 3A expression increases and inhibits oligodendrocyte precursor cell differentiation. Therefore, the remyelination of axons gets impaired. We hypothesized that the function of Sema 3A could be inhibited by neutralizing its binding to membrane NRP1. Methods we cloned a soluble form of mouse Neuropilin 1 (msNRP1) in a lentiviral vector and expressed the recombinant protein in HEK293T cells. Then, the conditioned medium of the transduced cells was used to evaluate the effects of the msNRP1 on the inhibition of Sema 3A-induced growth cone collapse activity. Dorsal root ganglion explants of timed pregnant (E13) mice were prepared. Then, the growth cone collapse activity of Sema 3A was assessed in the presence and absence of msNRP1-containing conditioned media of transduced and non-transduced HEK293T cells. Comparisons between groups were performed by 1-way ANOVA and post hoc Tukey tests. Results msNRP1 was successfully cloned and transduced in HEK293T cells. The supernatant of transduced cells was concentrated and evaluated for the production of msNRP1. ELISA results indicated that transduced cells secreted msNRP1. Growth cone collapse assay showed that Sema 3A activity was significantly reduced in the presence of the conditioned medium of msNRP1-transduced HEK293T cells. Conversely, a conditioned medium of non-transduced HEK293T cells could not effectively prevent Sema 3A growth cone collapse activity. Conclusion Our results indicated that msNRP1 was successfully produced in HEK293T cells. The secreted msNRP1 effectively prevented Sema 3A collapse activity. Therefore, msNRP1 can increase remyelination in MS lesions, although more studies using animal models are required.
Collapse
Affiliation(s)
- Keivan Nedaei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeideh Mazloomzadeh
- Social Determinants of Health Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehdi Totonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ali Reza Biglari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
4
|
Soleilhavoup C, Travaglio M, Patrick K, Garção P, Boobalan E, Adolfs Y, Spriggs RV, Moles-Garcia E, Dhiraj D, Oosterveen T, Ferri SL, Abel T, Brodkin ES, Pasterkamp RJ, Brooks BP, Panman L. Nolz1 expression is required in dopaminergic axon guidance and striatal innervation. Nat Commun 2020; 11:3111. [PMID: 32561725 PMCID: PMC7305235 DOI: 10.1038/s41467-020-16947-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 05/29/2020] [Indexed: 11/24/2022] Open
Abstract
Midbrain dopaminergic (DA) axons make long longitudinal projections towards the striatum. Despite the importance of DA striatal innervation, processes involved in establishment of DA axonal connectivity remain largely unknown. Here we demonstrate a striatal-specific requirement of transcriptional regulator Nolz1 in establishing DA circuitry formation. DA projections are misguided and fail to innervate the striatum in both constitutive and striatal-specific Nolz1 mutant embryos. The lack of striatal Nolz1 expression results in nigral to pallidal lineage conversion of striatal projection neuron subtypes. This lineage switch alters the composition of secreted factors influencing DA axonal tract formation and renders the striatum non-permissive for dopaminergic and other forebrain tracts. Furthermore, transcriptomic analysis of wild-type and Nolz1-/- mutant striatal tissue led to the identification of several secreted factors that underlie the observed guidance defects and proteins that promote DA axonal outgrowth. Together, our data demonstrate the involvement of the striatum in orchestrating dopaminergic circuitry formation.
Collapse
Affiliation(s)
- Clement Soleilhavoup
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Marco Travaglio
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Kieran Patrick
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Pedro Garção
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Elangovan Boobalan
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Ruth V Spriggs
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Emma Moles-Garcia
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Dalbir Dhiraj
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Tony Oosterveen
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Sarah L Ferri
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Edward S Brodkin
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104-3403, USA
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Brian P Brooks
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lia Panman
- MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK.
| |
Collapse
|
5
|
He Z, Crenshaw E, Raper JA. Semaphorin/neuropilin binding specificities are stable over 400 million years of evolution. Biochem Biophys Res Commun 2019; 517:23-28. [PMID: 31349972 PMCID: PMC10863626 DOI: 10.1016/j.bbrc.2019.06.133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 06/23/2019] [Indexed: 11/24/2022]
Abstract
Semaphorins are a large and important family of signaling molecules conserved in Bilateria. An important determinant of the biological function of their largest class, the secreted class 3 semaphorins, is the specificity of their binding to neuropilins, a key component of a larger holoreceptor complex. We compared these binding specificities in mice and zebrafish, species whose most recent common ancestor was more than 400 million years in the past. We also compared the binding specificities of zebrafish class 3 semaphorins that were duplicated very early within the teleost lineage. We found a surprising conservation of neuropilin binding specificities when comparing both paralogous zebrafish semaphorin pairs and orthologous zebrafish and mouse semaphorin pairs. This finding was further supported by a remarkable conservation of binding specificities in cross-species pairings of semaphorins and neuropilins. Our results suggest that the qualitative specificities with which particular semaphorins bind to particular neuropilins has remained nearly invariant over approximately 400 million years of evolution.
Collapse
Affiliation(s)
- Zhili He
- Dept. of Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ezekiel Crenshaw
- Dept. of Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan A Raper
- Dept. of Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Boggio EM, Ehlert EM, Lupori L, Moloney EB, De Winter F, Vander Kooi CW, Baroncelli L, Mecollari V, Blits B, Fawcett JW, Verhaagen J, Pizzorusso T. Inhibition of Semaphorin3A Promotes Ocular Dominance Plasticity in the Adult Rat Visual Cortex. Mol Neurobiol 2019; 56:5987-5997. [PMID: 30706367 DOI: 10.1007/s12035-019-1499-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 01/17/2019] [Indexed: 10/27/2022]
Abstract
Perineuronal nets (PNNs) are condensed structures in the extracellular matrix that mainly surround GABA-ergic parvalbumin-positive interneurons in the adult brain. Previous studies revealed a parallel between PNN formation and the closure of the critical period. Moreover, ocular dominance plasticity is enhanced in response to PNN manipulations in adult animals. However, the mechanisms through which perineuronal nets modulate plasticity are still poorly understood. Recent work indicated that perineuronal nets may convey molecular signals by binding and storing proteins with important roles in cellular communication. Here we report that semaphorin3A (Sema3A), a chemorepulsive axon guidance cue known to bind to important perineuronal net components, is necessary to dampen ocular dominance plasticity in adult rats. First, we showed that the accumulation of Sema3A in PNNs in the visual cortex correlates with critical period closure, following the same time course of perineuronal nets maturation. Second, the accumulation of Sema3A in perineuronal nets was significantly reduced by rearing animals in the dark in the absence of any visual experience. Finally, we developed and characterized a tool to interfere with Sema3A signaling by means of AAV-mediated expression of receptor bodies, soluble proteins formed by the extracellular domain of the endogenous Sema3A receptor (neuropilin1) fused to a human IgG Fc fragment. By using this tool to antagonize Sema3A signaling in the adult rat visual cortex, we found that the specific inhibition of Sema3A promoted ocular dominance plasticity. Thus, Sema3A accumulates in perineuronal nets in an experience-dependent manner and its presence in the mature visual cortex inhibits plasticity.
Collapse
Affiliation(s)
- Elena Maria Boggio
- Institute of Neuroscience, National Research Council CNR, Via Moruzzi, 1, 56124, Pisa, Italy
| | - Erich M Ehlert
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Leonardo Lupori
- BIO@SNS lab, Scuola Normale Superiore via Moruzzi, 1, 56124, Pisa, Italy
| | - Elizabeth B Moloney
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Fred De Winter
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Craig W Vander Kooi
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Laura Baroncelli
- Institute of Neuroscience, National Research Council CNR, Via Moruzzi, 1, 56124, Pisa, Italy
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128, Pisa, Italy
| | - Vasilis Mecollari
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Bas Blits
- UniQure, Meibergdreef 61, 1105 BA, Amsterdam, The Netherlands
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Robinson Way, Cambridge, CB2 0PY, UK
| | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 20 1085, 1081 HV, Amsterdam, The Netherlands
| | - Tommaso Pizzorusso
- Institute of Neuroscience, National Research Council CNR, Via Moruzzi, 1, 56124, Pisa, Italy.
- BIO@SNS lab, Scuola Normale Superiore via Moruzzi, 1, 56124, Pisa, Italy.
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA University of Florence, Area San Salvi - Pad. 26, 50135, Florence, Italy.
| |
Collapse
|
7
|
Xu J, Xu M, Wang Y, Mathena RP, Wen J, Zhang P, Furmanski O, Mintz CD. Anesthetics disrupt growth cone guidance cue sensing through actions on the GABA A α2 receptor mediated by the immature chloride gradient. Neurotoxicol Teratol 2019; 74:106812. [PMID: 31251980 DOI: 10.1016/j.ntt.2019.106812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/25/2019] [Accepted: 06/24/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND General anesthetics (GAs) may exert harmful effects on the developing brain by disrupting neuronal circuit formation. Anesthetics that act on γ-aminobutyric acid (GABA) receptors can interfere with axonal growth cone guidance, a critical process in the assembly of neuronal circuitry. Here we investigate the mechanism by which isoflurane prevents sensing of the repulsive guidance cue, Semaphorin 3A (Sema3A). METHODS Growth cone sensing was assayed by measuring growth cone collapse in dissociated neocortical cultures exposed to recombinant Sema3A in the presence or absence of isoflurane and/or a panel of reagents with specific actions on components of the GABA receptor and chloride ion systems. RESULTS Isoflurane exposure prevents Sema3A induced growth cone collapse. A GABAA α2 specific agonist replicates this effect (36.83 ± 3.417% vs 70.82 ± 2.941%, in the Sema3A induced control group, p < 0.0001), but an α1-specific agonist does not. Both a Na-K-Cl cotransporter 1 antagonism (bumetanide, BUM) and a chloride ionophore (IONO) prevent isoflurane from disrupting growth cone sensing of Sema3A. (65.67 ± 3.775% in Iso + BUM group vs 67.45 ± 3.624% in Sema3A induced control group, 65.34 ± 1.678% in Iso + IONO group vs 68.71 ± 2.071% in Sema3A induced control group, no significant difference) (n = 96 growth cones per group). CONCLUSION Our data suggest that the effects of isoflurane on growth cone sensing are mediated by the α2 subunit of the GABAA receptor and also that they are dependent on the developmental chloride gradient, in which Cl- exhibits a depolarizing effect. These findings provide a rationale for why immature neurons are particularly susceptible to anesthetic toxicity.
Collapse
Affiliation(s)
- Jing Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, 710004, China; Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Xu
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - YuChia Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - R Paige Mathena
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jieqiong Wen
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, 710004, China; Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, 710004, China
| | - Orion Furmanski
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - C David Mintz
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
8
|
Ryu JR, Kim JH, Cho HM, Jo Y, Lee B, Joo S, Chae U, Nam Y, Cho IJ, Sun W. A monitoring system for axonal growth dynamics using micropatterns of permissive and Semaphorin 3F chemorepulsive signals. LAB ON A CHIP 2019; 19:291-305. [PMID: 30539180 DOI: 10.1039/c8lc00845k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Neurons reach their correct targets by directional outgrowth of axons, which is mediated by attractive or repulsive cues. Growing axons occasionally cross a field of repulsive cues and stop at intermediate targets on the journey to their final destination. However, it is not well-understood how individual growth cones make decisions, and pass through repulsive territory to reach their permissive target regions. We developed a microcontact printing culture system that could trap individual axonal tips in a permissive dot area surrounded by the repulsive signal, semaphorin 3F (Sema3F). Axons of rat hippocampal neurons on the Sema3F/PLL dot array extended in the checkboard pattern with a significantly slow growth rate. The detailed analysis of the behaviors of axonal growth cones revealed the saccadic dynamics in the dot array system. The trapped axonal tips in the permissive area underwent growth cone enlargement with remarkably spiky filopodia, promoting their escape from the Sema3F constraints with straight extension of axons. This structured axonal growth on the dot pattern was disrupted by increased inter-dot distance, or perturbing intracellular signaling machineries. These data indicate that axons grow against repulsive signals by jumping over the repulsive cues, depending on contact signals and intracellular milieu. Our study suggests that our dot array culture system can be used as a screening system to easily and efficiently evaluate ECM or small molecule inhibitors interfering growth cone dynamics leading to controlling axonal growth.
Collapse
Affiliation(s)
- Jae Ryun Ryu
- Department of Anatomy, Brain Korea 21, Korea University College of Medicine, Anam-Dong, Sungbuk-Gu, Seoul, 136-705, Republic of Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
The soluble form of LOTUS inhibits Nogo receptor type 1-mediated signaling induced by B lymphocyte stimulator and chondroitin sulfate proteoglycans. Neurosci Lett 2018; 683:61-68. [PMID: 29953923 DOI: 10.1016/j.neulet.2018.06.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 12/12/2022]
Abstract
There are global efforts in developing therapeutic strategies for central nervous system (CNS) injuries using multimodal approaches. Nogo receptor type 1 (NgR1) has been known as a primary molecule limiting neuronal regeneration in the adult CNS. We identified lateral olfactory tract usher substance (LOTUS) as an endogenous NgR1 antagonist. Membrane-bound LOTUS interacts with NgR1 and inhibits its function by blocking its ligand binding. Five molecules including Nogo, myelin-associated glycoprotein (MAG), oligodendrocyte myelin glycoprotein (OMgp), B lymphocyte stimulator (BLyS) and chondroitin sulfate proteoglycans (CSPGs) have been identified as NgR1 ligands. These ligands bind to NgR1 and activate NgR1 signaling, leading to axon growth inhibition such as growth cone collapse and neurite outgrowth inhibition. We have recently reported that the soluble form of LOTUS (s-LOTUS) also suppressed NgR1-mediated signaling induced by myelin axonal inhibitors (MAIs) including Nogo, MAG and OMgp by binding with both NgR1 and its co-receptor p75 neurotrophin receptor (p75NTR). Though s-LOTUS has been reported to suppress MAIs, whether s-LOTUS also suppresses NgR1 signaling induced by BLyS and CSPGs remains to be elucidated. Here, we show that s-LOTUS inhibits NgR1-mediated signaling induced by BLyS and CSPGs. Although treatment with s-LOTUS did not suppress BLyS-NgR1 interaction, s-LOTUS inhibited growth cone collapse and neurite outgrowth inhibition induced by BLyS and CSPGs in chick dorsal root ganglion (DRG) neurons. Furthermore, s-LOTUS compensated for the suppressive function of endogenous LOTUS in NgR1-mediated signaling in olfactory bulb neurons of lotus-knockout mice. These findings suggest that s-LOTUS is a potent therapeutic agent for neuronal regeneration in the CNS injuries.
Collapse
|
10
|
Schaeffer J, Tannahill D, Cioni JM, Rowlands D, Keynes R. Identification of the extracellular matrix protein Fibulin-2 as a regulator of spinal nerve organization. Dev Biol 2018; 442:101-114. [PMID: 29944871 DOI: 10.1016/j.ydbio.2018.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 05/24/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022]
Abstract
During amniote peripheral nervous system development, segmentation ensures the correct patterning of the spinal nerves relative to the vertebral column. Along the antero-posterior (rostro-caudal) axis, each somite-derived posterior half-sclerotome expresses repellent molecules to restrict axon growth and neural crest migration to the permissive anterior half-segment. To identify novel regulators of spinal nerve patterning, we investigated the differential gene expression of anterior and posterior half-sclerotomes in the chick embryo by RNA-sequencing. Several genes encoding extracellular matrix proteins were found to be enriched in either anterior (e.g. Tenascin-C, Laminin alpha 4) or posterior (e.g. Fibulin-2, Fibromodulin, Collagen VI alpha 2) half-sclerotomes. Among them, the extracellular matrix protein Fibulin-2 was found specifically restricted to the posterior half-sclerotome. By using in ovo ectopic expression in chick somites, we found that Fibulin-2 modulates spinal axon growth trajectories in vivo. While no intrinsic axon repellent activity of Fibulin-2 was found, we showed that it enhances the growth cone repulsive activity of Semaphorin 3A in vitro. Some molecules regulating axon growth during development are found to be upregulated in the adult central nervous system (CNS) following traumatic injury. Here, we found increased Fibulin-2 protein levels in reactive astrocytes at the lesion site of a mouse model of CNS injury. Together, these results suggest that the developing vertebral column and the adult CNS share molecular features that control axon growth and plasticity, which may open up the possibility for the identification of novel therapeutic targets for brain and spinal cord injury.
Collapse
Affiliation(s)
- Julia Schaeffer
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK.
| | | | - Jean-Michel Cioni
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | | | - Roger Keynes
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| |
Collapse
|
11
|
Kawakami Y, Kurihara Y, Saito Y, Fujita Y, Yamashita T, Takei K. The Soluble Form of LOTUS inhibits Nogo Receptor-Mediated Signaling by Interfering with the Interaction Between Nogo Receptor Type 1 and p75 Neurotrophin Receptor. J Neurosci 2018; 38:2589-2604. [PMID: 29440387 PMCID: PMC6705898 DOI: 10.1523/jneurosci.0953-17.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 01/25/2023] Open
Abstract
Nogo receptor type 1 (NgR1) is known to inhibit neuronal regeneration in the CNS. Previously, we have shown that lateral olfactory tract usher substance (LOTUS) interacts with NgR1 and inhibits its function by blocking its ligand binding. Therefore, LOTUS is expected to have therapeutic potential for the promotion of neuronal regeneration. However, it remains unknown whether the soluble form of LOTUS (s-LOTUS) also has an inhibitory action on NgR1 function as a candidate for therapeutic agents. Here, we show that s-LOTUS inhibits NgR1-mediated signaling by inhibiting the molecular interaction between NgR1 and its coreceptor, p75 neurotrophin receptor (p75NTR). In contrast to the membrane-bound form of LOTUS, s-LOTUS did not block ligand binding to NgR1. However, we identified p75NTR as a novel LOTUS binding partner and found that s-LOTUS suppressed the interaction between p75NTR and NgR1. s-LOTUS inhibited myelin-associated inhibitor (MAI)-induced RhoA activation in murine cortical neurons. Functional analyses revealed that s-LOTUS inhibited MAI-induced growth cone collapse and neurite outgrowth inhibition in chick DRG neurons. In addition, whereas olfactory bulb neurons of lotus-KO mice are sensitive to MAI due to a lack of LOTUS expression, treatment with s-LOTUS inhibited MAI-induced growth cone collapse in these neurons. Finally, we observed that s-LOTUS promoted axonal regeneration in optic nerve crush injury of mice (either sex). These findings suggest that s-LOTUS inhibits NgR1-mediated signaling, possibly by interfering with the interaction between NgR1 and p75NTR Therefore, s-LOTUS may have potential as a therapeutic agent for neuronal regeneration in the damaged CNS.SIGNIFICANCE STATEMENT Nogo receptor type 1 (NgR1) is a receptor well known to inhibit neuronal regeneration in the CNS. Because the membrane-bound form of lateral olfactory tract usher substance (LOTUS) antagonizes NgR1 through a cis-type molecular interaction between LOTUS and NgR1, the soluble form of LOTUS (s-LOTUS) is expected to be a therapeutic agent for neuronal regeneration. In our present study, we show that s-LOTUS inhibits the interaction between NgR1 and p75NTR, NgR1 ligand-induced RhoA activation, growth cone collapse, and neurite outgrowth inhibition and promotes axonal regeneration. Our results indicate that s-LOTUS inhibits NgR1-mediated signaling through a trans-type molecular interaction between LOTUS and NgR1 and, therefore, s-LOTUS may have therapeutic potential for neuronal regeneration.
Collapse
Affiliation(s)
- Yutaka Kawakami
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan and
| | - Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan and
| | - Yu Saito
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan and
| | - Yuki Fujita
- Department of Molecular Neuroscience, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan and
| |
Collapse
|
12
|
Fiederling F, Weschenfelder M, Fritz M, von Philipsborn A, Bastmeyer M, Weth F. Ephrin-A/EphA specific co-adaptation as a novel mechanism in topographic axon guidance. eLife 2017; 6. [PMID: 28722651 PMCID: PMC5517148 DOI: 10.7554/elife.25533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/26/2017] [Indexed: 12/30/2022] Open
Abstract
Genetic hardwiring during brain development provides computational architectures for innate neuronal processing. Thus, the paradigmatic chick retinotectal projection, due to its neighborhood preserving, topographic organization, establishes millions of parallel channels for incremental visual field analysis. Retinal axons receive targeting information from quantitative guidance cue gradients. Surprisingly, novel adaptation assays demonstrate that retinal growth cones robustly adapt towards ephrin-A/EphA forward and reverse signals, which provide the major mapping cues. Computational modeling suggests that topographic accuracy and adaptability, though seemingly incompatible, could be reconciled by a novel mechanism of coupled adaptation of signaling channels. Experimentally, we find such 'co-adaptation' in retinal growth cones specifically for ephrin-A/EphA signaling. Co-adaptation involves trafficking of unliganded sensors between the surface membrane and recycling endosomes, and is presumably triggered by changes in the lipid composition of membrane microdomains. We propose that co-adaptative desensitization eventually relies on guidance sensor translocation into cis-signaling endosomes to outbalance repulsive trans-signaling.
Collapse
Affiliation(s)
- Felix Fiederling
- Department of Cell and Neurobiology, Karlsruhe Institute of Technology, Zoological Institute, Karlruhe, Germany
| | - Markus Weschenfelder
- Department of Cell and Neurobiology, Karlsruhe Institute of Technology, Zoological Institute, Karlruhe, Germany
| | - Martin Fritz
- Department of Cell and Neurobiology, Karlsruhe Institute of Technology, Zoological Institute, Karlruhe, Germany
| | - Anne von Philipsborn
- Department of Cell and Neurobiology, Karlsruhe Institute of Technology, Zoological Institute, Karlruhe, Germany
| | - Martin Bastmeyer
- Department of Cell and Neurobiology, Karlsruhe Institute of Technology, Zoological Institute, Karlruhe, Germany
| | - Franco Weth
- Department of Cell and Neurobiology, Karlsruhe Institute of Technology, Zoological Institute, Karlruhe, Germany
| |
Collapse
|
13
|
Kurihara Y, Saito Y, Takei K. Blockade of chondroitin sulfate proteoglycans-induced axonal growth inhibition by LOTUS. Neuroscience 2017; 356:265-274. [PMID: 28571719 DOI: 10.1016/j.neuroscience.2017.05.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/14/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are axon growth inhibitors in the glial scar, and restrict axon regeneration following damage to the adult mammalian central nervous system. CSPGs have recently been identified as functional ligands for Nogo receptor-1 (NgR1), which is the common receptor for Nogo proteins, myelin-associated glycoprotein (MAG), oligodendrocyte myelin glycoprotein (OMgp) and B lymphocyte stimulator (BLyS). We have previously reported that through its binding to NgR1, lateral olfactory tract usher substance (LOTUS) suppresses Nogo, MAG, OMgp, and BLyS-induced axon growth inhibition. However, it remains unknown whether LOTUS also exerts this suppressive action on CSPG-induced axon growth inhibition. LOTUS overexpression rescued CSPG-induced growth cone collapse and neurite outgrowth inhibition in cultured dorsal root ganglion neurons, which only weakly express endogenous LOTUS. In cultured olfactory bulb neurons, which endogenously express LOTUS, the growth cone was insensitive to CSPG-induced collapse, but was sensitive to collapse induced by CSPGs in lotus-deficient mice. Our data demonstrate that LOTUS suppresses CSPG-induced axon growth inhibition, suggesting that LOTUS may represent a promising therapeutic agent for promoting axon regeneration.
Collapse
Affiliation(s)
- Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Yu Saito
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan.
| |
Collapse
|
14
|
Using Rotary Shadow Electron Microscopy to Characterize Semaphorin-Mediated Growth Cone Collapse. Methods Mol Biol 2016. [PMID: 27787851 DOI: 10.1007/978-1-4939-6448-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Rotary shadow electron microscopy (EM) of growth cone cytoskeletons provides a high-resolution method for detecting both global and macromolecular changes in cytoskeletal organization or structure. This approach can be used to study responses to repulsive guidance factors such as semaphorin 3A. Here I describe the procedures used to prepare cultured neurons for rotary-shadow EM, allowing detailed comparisons of cytoskeletal structure.
Collapse
|
15
|
Ahmed YA, Yates EA, Moss DJ, Loeven MA, Hussain SA, Hohenester E, Turnbull JE, Powell AK. Panels of chemically-modified heparin polysaccharides and natural heparan sulfate saccharides both exhibit differences in binding to Slit and Robo, as well as variation between protein binding and cellular activity. MOLECULAR BIOSYSTEMS 2016; 12:3166-75. [PMID: 27502551 PMCID: PMC5048398 DOI: 10.1039/c6mb00432f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/04/2016] [Indexed: 01/09/2023]
Abstract
Heparin/heparan sulfate (HS) glycosaminoglycans are required for Slit-Robo cellular responses. Evidence exists for interactions between each combination of Slit, Robo and heparin/HS and for formation of a ternary complex. Heparin/HS are complex mixtures displaying extensive structural diversity. The relevance of this diversity has been studied to a limited extent using a few select chemically-modified heparins as models of HS diversity. Here we extend these studies by parallel screening of structurally diverse panels of eight chemically-modified heparin polysaccharides and numerous natural HS oligosaccharide chromatographic fractions for binding to both Drosophila Slit and Robo N-terminal domains and for activation of a chick retina axon response to the Slit fragment. Both the polysaccharides and oligosaccharide fractions displayed variability in binding and cellular activity that could not be attributed solely to increasing sulfation, extending evidence for the importance of structural diversity to natural HS as well as model modified heparins. They also displayed differences in their interactions with Slit compared to Robo, with Robo preferring compounds with higher sulfation. Furthermore, the patterns of cellular activity across compounds were different to those for binding to each protein, suggesting that biological outcomes are selectively determined in a subtle manner that does not simply reflect the sum of the separate interactions of heparin/HS with Slit and Robo.
Collapse
Affiliation(s)
- Yassir A. Ahmed
- Centre for Glycobiology , Institute of Integrative Biology , University of Liverpool , UK
- Department of Chemistry , Faculty of Science , King Faisal University , Kingdom of Saudi Arabia
| | - Edwin A. Yates
- Centre for Glycobiology , Institute of Integrative Biology , University of Liverpool , UK
| | - Diana J. Moss
- Department of Cellular and Molecular Physiology , University of Liverpool , UK
| | - Markus A. Loeven
- Centre for Glycobiology , Institute of Integrative Biology , University of Liverpool , UK
| | | | | | - Jeremy E. Turnbull
- Centre for Glycobiology , Institute of Integrative Biology , University of Liverpool , UK
| | - Andrew K. Powell
- Centre for Glycobiology , Institute of Integrative Biology , University of Liverpool , UK
- School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , UK .
| |
Collapse
|
16
|
Repulsive axon guidance by Draxin is mediated by protein Kinase B (Akt), glycogen synthase kinase-3β (GSK-3β) and microtubule-associated protein 1B. PLoS One 2015; 10:e0119524. [PMID: 25775433 PMCID: PMC4361590 DOI: 10.1371/journal.pone.0119524] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/13/2015] [Indexed: 11/19/2022] Open
Abstract
Draxin is an important axon guidance cue necessary for the formation of forebrain commissures including the corpus callosum, but the molecular details of draxin signaling are unknown. To unravel how draxin signals are propagated we used murine cortical neurons and genetic and pharmacological approaches. We found that draxin-induced growth cone collapse critically depends on draxin receptors (deleted in colorectal cancer, DCC), inhibition of protein kinase B/Akt, activation of GSK-3β (glycogen synthase kinase-3β) and the presence of microtubule-associated protein MAP1B. This study, for the first time elucidates molecular events in draxin repulsion, links draxin and DCC to MAP1B and identifies a novel MAP1B-depenent GSK-3β pathway essential for chemo-repulsive axon guidance cue signaling.
Collapse
|
17
|
Sharma A, LeVaillant CJ, Plant GW, Harvey AR. Changes in expression of Class 3 Semaphorins and their receptors during development of the rat retina and superior colliculus. BMC DEVELOPMENTAL BIOLOGY 2014; 14:34. [PMID: 25062604 PMCID: PMC4121511 DOI: 10.1186/s12861-014-0034-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 07/15/2014] [Indexed: 12/28/2022]
Abstract
Background Members of the Semaphorin 3 family (Sema3s) influence the development of the central nervous system, and some are implicated in regulating aspects of visual system development. However, we lack information about the timing of expression of the Sema3s with respect to different developmental epochs in the mammalian visual system. In this time-course study in the rat, we document for the first time changes in the expression of RNAs for the majority of Class 3 Semaphorins (Sema3s) and their receptor components during the development of the rat retina and superior colliculus (SC). Results During retinal development, transcript levels changed for all of the Sema3s examined, as well as Nrp2, Plxna2, Plxna3, and Plxna4a. In the SC there were also changes in transcript levels for all Sema3s tested, as well as Nrp1, Nrp2, Plxna1, Plxna2, Plxna3, and Plxna4a. These changes correlate with well-established epochs, and our data suggest that the Sema3s could influence retinal ganglion cell (RGC) apoptosis, patterning and connectivity in the maturing retina and SC, and perhaps guidance of RGC and cortical axons in the SC. Functionally we found that SEMA3A, SEMA3C, SEMA3E, and SEMA3F proteins collapsed purified postnatal day 1 RGC growth cones in vitro. Significantly this is a developmental stage when RGCs are growing into and within the SC and are exposed to Sema3 ligands. Conclusion These new data describing the overall temporal regulation of Sema3 expression in the rat retina and SC provide a platform for further work characterising the functional impact of these proteins on the development and maturation of mammalian visual pathways.
Collapse
Affiliation(s)
- Anil Sharma
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley 6009, WA, Australia.
| | | | | | | |
Collapse
|
18
|
Kurihara Y, Iketani M, Ito H, Nishiyama K, Sakakibara Y, Goshima Y, Takei K. LOTUS suppresses axon growth inhibition by blocking interaction between Nogo receptor-1 and all four types of its ligand. Mol Cell Neurosci 2014; 61:211-8. [PMID: 25034269 DOI: 10.1016/j.mcn.2014.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 10/25/2022] Open
Abstract
Axon growth inhibitors such as Nogo proteins, myelin-associated glycoprotein (MAG), oligodendrocyte myelin glycoprotein (OMgp), and B lymphocyte stimulator (BLyS) commonly bind to Nogo receptor-1 (NgR1), leading to enormous restriction of functional recovery after damage to the adult central nervous system. Recently, we found that lateral olfactory tract usher substance (LOTUS) antagonizes NgR1-mediated Nogo signaling. However, whether LOTUS exerts antagonism of NgR1 when bound by the other three ligands has not been determined. Overexpression of LOTUS together with NgR1 in COS7 cells blocked the binding of MAG, OMgp, and BLyS to NgR1. In cultured dorsal root ganglion neurons in which endogenous LOTUS is only weakly expressed, overexpression of LOTUS suppressed growth cone collapse and neurite outgrowth inhibition induced by these three NgR1 ligands. LOTUS suppressed NgR1 ligand-induced growth cone collapse in cultured olfactory bulb neurons, which endogenously express LOTUS. Growth cone collapse was induced by NgR1 ligands in lotus-deficient mice. These data suggest that LOTUS functions as a potent endogenous antagonist for NgR1 when bound by all four known NgR1 ligands, raising the possibility that LOTUS may protect neurons from NgR1-mediated axonal growth inhibition and thereby may be useful for promoting neuronal regeneration as a potent inhibitor of NgR1.
Collapse
Affiliation(s)
- Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Masumi Iketani
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Hiromu Ito
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ward, Yokohama 236-0004, Japan
| | - Kuniyuki Nishiyama
- Division of Medical Life Science, Yokohama City University School of Medicine, Fukuura 3-9, Kanazawa-ward, Yokohama 236-0004, Japan; Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ward, Yokohama 236-0004, Japan
| | - Yusuke Sakakibara
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ward, Yokohama 236-0004, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ward, Yokohama 236-0004, Japan; Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ward, Yokohama 236-0004, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan; Division of Medical Life Science, Yokohama City University School of Medicine, Fukuura 3-9, Kanazawa-ward, Yokohama 236-0004, Japan; Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ward, Yokohama 236-0004, Japan.
| |
Collapse
|
19
|
Abstract
The growth cone collapse assay has proved invaluable in detecting and purifying axonal repellents. Glycoproteins/proteins present in detergent extracts of biological tissues are incorporated into liposomes, added to growth cones in culture and changes in morphology are then assessed. Alternatively purified or recombinant molecules in aqueous solution may be added directly to the cultures. In both cases after a defined period of time (up to 1 h), the cultures are fixed and then assessed by inverted phase contrast microscopy for the percentage of growth cones showing a collapsed profile with loss of flattened morphology, filopodia, and lamellipodia.
Collapse
|
20
|
Häbig K, Gellhaar S, Heim B, Djuric V, Giesert F, Wurst W, Walter C, Hentrich T, Riess O, Bonin M. LRRK2 guides the actin cytoskeleton at growth cones together with ARHGEF7 and Tropomyosin 4. Biochim Biophys Acta Mol Basis Dis 2013; 1832:2352-67. [DOI: 10.1016/j.bbadis.2013.09.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 08/06/2013] [Accepted: 09/16/2013] [Indexed: 11/27/2022]
|
21
|
Tran TS, Carlin E, Lin R, Martinez E, Johnson JE, Kaprielian Z. Neuropilin2 regulates the guidance of post-crossing spinal commissural axons in a subtype-specific manner. Neural Dev 2013; 8:15. [PMID: 23902858 PMCID: PMC3737016 DOI: 10.1186/1749-8104-8-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/19/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Spinal commissural axons represent a model system for deciphering the molecular logic that regulates the guidance of midline-crossing axons in the developing central nervous system (CNS). Whether the same or specific sets of guidance signals control the navigation of molecularly distinct subtypes of these axons remains an open and largely unexplored question. Although it is well established that post-crossing commissural axons alter their responsiveness to midline-associated guidance cues, our understanding of the repulsive mechanisms that drive the post-crossing segments of these axons away from the midline and whether the underlying guidance systems operate in a commissural axon subtype-specific manner, remains fragmentary at best. RESULTS Here, we utilize axonally targeted transgenic reporter mice to visualize genetically distinct dorsal interneuron (dI)1 and dI4 commissural axons and show that the repulsive class 3 semaphorin (Sema3) guidance receptor Neuropilin 2 (Npn2), is selectively expressed on the dI1 population and is required for the guidance of post-crossing dI1, but not dI4, axons. Consistent with these observations, the midline-associated Npn2 ligands, Sema3F and Sema3B, promote the collapse of dI1, but not dI4, axon-associated growth cones in vitro. We also identify, for the first time, a discrete GABAergic population of ventral commissural neurons/axons in the embryonic mouse spinal cord that expresses Npn2, and show that Npn2 is required for the proper guidance of their post-crossing axons. CONCLUSIONS Together, our findings indicate that Npn2 is selectively expressed in distinct populations of commissural neurons in both the dorsal and ventral spinal cord, and suggest that Sema3-Npn2 signaling regulates the guidance of post-crossing commissural axons in a population-specific manner.
Collapse
Affiliation(s)
- Tracy S Tran
- Department of Biological Sciences, Rutgers University, Boyden 206, 195 University Ave,, Newark, NJ 07102, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Anesthetics interfere with axon guidance in developing mouse neocortical neurons in vitro via a γ-aminobutyric acid type A receptor mechanism. Anesthesiology 2013; 118:825-33. [PMID: 23364597 DOI: 10.1097/aln.0b013e318287b850] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The finding that exposure to general anesthetics (GAs) in childhood may increase rates of learning disabilities has raised a concern that anesthetics may interfere with brain development. The generation of neuronal circuits, a complex process in which axons follow guidance cues to dendritic targets, is an unexplored potential target for this type of toxicity. METHODS GA exposures were conducted in developing neocortical neurons in culture and in early postnatal neocortical slices overlaid with fluorescently labeled neurons. Axon targeting, growth cone collapse, and axon branching were measured using quantitative fluorescence microscopy. RESULTS Isoflurane exposure causes errors in Semaphorin-3A-dependent axon targeting (n = 77 axons) and a disruption of the response of axonal growth cones to Semaphorin-3A (n = 2,358 growth cones). This effect occurs at clinically relevant anesthetic doses of numerous GAs with allosteric activity at γ-aminobutyric acid type A receptors, and it was reproduced with a selective agonist. Isoflurane also inhibits growth cone collapse induced by Netrin-1, but does not interfere branch induction by Netrin-1. Insensitivity to guidance cues caused by isoflurane is seen acutely in growth cones in dissociated culture, and errors in axon targeting in brain slice culture occur at the earliest point at which correct targeting is observed in controls. CONCLUSIONS These results demonstrate a generalized inhibitory effect of GAs on repulsive growth cone guidance in the developing neocortex that may occur via a γ-aminobutyric acid type A receptor mechanism. The finding that GAs interfere with axon guidance, and thus potentially with circuit formation, represents a novel form of anesthesia neurotoxicity in brain development.
Collapse
|
23
|
Nakano Y, Chayama K, Ochi H, Toshishige M, Hayashida Y, Miki D, Hayes CN, Suzuki H, Tokuyama T, Oda N, Suenari K, Uchimura-Makita Y, Kajihara K, Sairaku A, Motoda C, Fujiwara M, Watanabe Y, Yoshida Y, Ohkubo K, Watanabe I, Nogami A, Hasegawa K, Watanabe H, Endo N, Aiba T, Shimizu W, Ohno S, Horie M, Arihiro K, Tashiro S, Makita N, Kihara Y. A nonsynonymous polymorphism in semaphorin 3A as a risk factor for human unexplained cardiac arrest with documented ventricular fibrillation. PLoS Genet 2013; 9:e1003364. [PMID: 23593010 PMCID: PMC3623806 DOI: 10.1371/journal.pgen.1003364] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 01/19/2013] [Indexed: 11/19/2022] Open
Abstract
Unexplained cardiac arrest (UCA) with documented ventricular fibrillation (VF) is a major cause of sudden cardiac death. Abnormal sympathetic innervations have been shown to be a trigger of ventricular fibrillation. Further, adequate expression of SEMA3A was reported to be critical for normal patterning of cardiac sympathetic innervation. We investigated the relevance of the semaphorin 3A (SEMA3A) gene located at chromosome 5 in the etiology of UCA. Eighty-three Japanese patients diagnosed with UCA and 2,958 healthy controls from two different geographic regions in Japan were enrolled. A nonsynonymous polymorphism (I334V, rs138694505A>G) in exon 10 of the SEMA3A gene identified through resequencing was significantly associated with UCA (combined P = 0.0004, OR 3.08, 95%CI 1.67-5.7). Overall, 15.7% of UCA patients carried the risk genotype G, whereas only 5.6% did in controls. In patients with SEMA3A(I334V), VF predominantly occurred at rest during the night. They showed sinus bradycardia, and their RR intervals on the 12-lead electrocardiography tended to be longer than those in patients without SEMA3A(I334V) (1031±111 ms versus 932±182 ms, P = 0.039). Immunofluorescence staining of cardiac biopsy specimens revealed that sympathetic nerves, which are absent in the subendocardial layer in normal hearts, extended to the subendocardial layer only in patients with SEMA3A(I334V). Functional analyses revealed that the axon-repelling and axon-collapsing activities of mutant SEMA3A(I334V) genes were significantly weaker than those of wild-type SEMA3A genes. A high incidence of SEMA3A(I334V) in UCA patients and inappropriate innervation patterning in their hearts implicate involvement of the SEMA3A gene in the pathogenesis of UCA.
Collapse
Affiliation(s)
- Yukiko Nakano
- Department of Cardiovascular Medicine, Division of Frontier Medical Science, Programs for Biomedical Research, Graduate School of Biomedical Science, Hiroshima University, Hiroshima, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Growth cone collapse is an easy and efficient test for detecting and characterizing axon guidance activities secreted or expressed by cells. It can also be used to dissect signaling pathways by axon growth inhibitors and to isolate therapeutic compounds that promote axon regeneration. Here, we describe a growth cone collapse assay protocol used to study signal transduction mechanisms of the repulsive axon guidance molecule ephrin-A5 in hippocampal neurons.
Collapse
Affiliation(s)
- Xin Yue
- Department of Chemical Biology, Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | | | | |
Collapse
|
25
|
Janssen BJ, Malinauskas T, Weir GA, Cader MZ, Siebold C, Jones EY. Neuropilins lock secreted semaphorins onto plexins in a ternary signaling complex. Nat Struct Mol Biol 2012; 19:1293-9. [PMID: 23104057 PMCID: PMC3590443 DOI: 10.1038/nsmb.2416] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 09/18/2012] [Indexed: 12/15/2022]
Abstract
Co-receptors add complexity to cell-cell signaling systems. The secreted semaphorin 3s (Sema3s) require a co-receptor, neuropilin (Nrp), to signal through plexin As (PlxnAs) in functions ranging from axon guidance to bone homeostasis, but the role of the co-receptor is obscure. Here we present the low-resolution crystal structure of a mouse semaphorin-plexin-Nrp complex alongside unliganded component structures. Dimeric semaphorin, two copies of plexin and two copies of Nrp are arranged as a dimer of heterotrimers. In each heterotrimer subcomplex, semaphorin contacts plexin, similar to in co-receptor-independent signaling complexes. The Nrp1s cross brace the assembly, bridging between sema domains of the Sema3A and PlxnA2 subunits from the two heterotrimers. Biophysical and cellular analyses confirm that this Nrp binding mode stabilizes a canonical, but weakened, Sema3-PlxnA interaction, adding co-receptor control over the mechanism by which receptor dimerization and/or oligomerization triggers signaling.
Collapse
Affiliation(s)
- Bert J.C. Janssen
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Greg A. Weir
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - M. Zameel Cader
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Christian Siebold
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - E. Yvonne Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
26
|
Sharma A, Verhaagen J, Harvey AR. Receptor complexes for each of the Class 3 Semaphorins. Front Cell Neurosci 2012; 6:28. [PMID: 22783168 PMCID: PMC3389612 DOI: 10.3389/fncel.2012.00028] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 06/20/2012] [Indexed: 01/08/2023] Open
Abstract
The Class 3 Semaphorins (Sema3s) are a sub-family of proteins whose known biological roles are varied and growing. The mechanism of action of the Sema3s requires binding to transmembrane receptors that comprise heteromeric complexes of Neuropilins, Plexins and cell adhesion molecules (CAMs). However, knowledge of the receptor components of the Sema3s remains incomplete, and there may be receptor components which are as yet undiscovered. The receptor complexes of the Sema3s share receptor components with each other, and it is the specific combination of these components within a heteromeric complex that is thought to give rise to selective binding and signalling for individual Sema3s. This crosstalk makes it experimentally difficult to define a single holoreceptor for each Sema3. Furthermore, the receptor composition for a given Sema3 may differ between cell types, and change as a function of developmental state or pathological situation. Nevertheless, there are at least some known differences in the constitutive structure of the receptors for the Sema3s. For example in neural cells, Sema3a and Sema3f signal through different Neuropilins (Nrp1 and Nrp2 respectively) and L1cam only appears important for Sema3a signaling, while Nrcam forms a complex with Nrp2. Further complexity arises from crosstalk of other families of ligands (e.g., VEGF) with Sema3 receptor components. Thus the Sema3s, which have been shown as antagonists for each other, can also act as antagonists for other families of molecules. This review compiles experimental evidence describing the receptor components for the Sema3s, detailing the current state of knowledge of which components are important for signaling of each Sema3 before going on to consider possible future directions for the field.
Collapse
Affiliation(s)
- Anil Sharma
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley WA, Australia
| | | | | |
Collapse
|
27
|
Zheng W, Geng AQ, Li PF, Wang Y, Yuan XB. Robo4 regulates the radial migration of newborn neurons in developing neocortex. ACTA ACUST UNITED AC 2011; 22:2587-601. [PMID: 22123939 DOI: 10.1093/cercor/bhr330] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During the morphogenesis of neocortex, newborn neurons undergo radial migration from the ventricular zone toward the surface of the cortical plate to form an "inside-out" lamina structure. The spatiotemporal signals that control this stereotyped radial migration remain elusive. Here, we report that a recently identified Robo family member Robo4 (Magic Roundabout), which was considered to be solely expressed in endothelial cells, is expressed in developing brain and regulates the radial migration of newborn neurons in neocortex. Downregulation of Robo4 expression in cortical newborn neurons by using in utero electroporation, with either specific siRNAs in wild-type rodents or with Cre recombinase in floxed-robo4 mutant mice, led to severe defects in the radial migration of newborn neurons with misorientation of these neurons. Moreover, newborn neurons transfected with Robo4 siRNAs exhibited significantly lower motility in a transwell migration assay (Boyden chamber) in the absence of Slit and significantly higher sensitivity to the repulsive effect of Slit in both transwell migration assay and growth cone collapse assay. Overall, our results showed an important role of Robo4 in the regulation of cortical radial migration through Slit-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Wang Zheng
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | |
Collapse
|
28
|
Morgan TE, Davis DA, Iwata N, Tanner JA, Snyder D, Ning Z, Kam W, Hsu YT, Winkler JW, Chen JC, Petasis NA, Baudry M, Sioutas C, Finch CE. Glutamatergic neurons in rodent models respond to nanoscale particulate urban air pollutants in vivo and in vitro. ENVIRONMENTAL HEALTH PERSPECTIVES 2011; 119:1003-9. [PMID: 21724521 PMCID: PMC3222976 DOI: 10.1289/ehp.1002973] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 03/29/2011] [Indexed: 05/06/2023]
Abstract
BACKGROUND Inhalation of airborne particulate matter (PM) derived from urban traffic is associated with pathology in the arteries, heart, and lung; effects on brain are also indicated but are less documented. OBJECTIVE We evaluated rodent brain responses to urban nanoscale (< 200 nm) PM (nPM). METHODS Ambient nPM collected near an urban freeway was transferred to aqueous suspension and reaerosolized for 10-week inhalation exposure of mice or directly applied to rat brain cell cultures. RESULTS Free radicals were detected by electron paramagnetic resonance in the nPM 30 days after initial collection. Chronic inhalation of reaerosolized nPM altered selected neuronal and glial activities in mice. The neuronal glutamate receptor subunit (GluA1) was decreased in hippocampus, whereas glia were activated and inflammatory cytokines were induced [interleukin-1α (IL-1α), tumor necrosis factor-α (TNFα)] in cerebral cortex. Two in vitro models showed effects of nPM suspensions within 24-48 hr of exposure that involved glutamatergic functions. In hippocampal slice cultures, nPM increased the neurotoxicity of NMDA (N-methyl-d-aspartic acid), a glutamatergic agonist, which was in turn blocked by the NMDA antagonist AP5 [(2R)-amino-5-phosphonopentanoate]. In embryonic neuron cultures, nPM impaired neurite outgrowth, also blocked by AP5. Induction of IL-1α and TNFα in mixed glia cultures required higher nPM concentrations than did neuronal effects. Because conditioned media from nPM-exposed glia also impaired outgrowth of embryonic neurites, nPM can act indirectly, as well as directly, on neurons in vitro. CONCLUSIONS nPM can affect embryonic and adult neurons through glutamatergic mechanisms. The interactions of nPM with glutamatergic neuronal functions suggest that cerebral ischemia, which involves glutamatergic excitotoxicity, could be exacerbated by nPM.
Collapse
Affiliation(s)
- Todd E Morgan
- Davis School of Gerontology, University of Southern California, Los Angeles, California 90089, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sepulveda B, Carcea I, Zhao B, Salton SR, Benson DL. L1 cell adhesion molecule promotes resistance to alcohol-induced silencing of growth cone responses to guidance cues. Neuroscience 2011; 180:30-40. [PMID: 21335065 PMCID: PMC3070798 DOI: 10.1016/j.neuroscience.2011.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 01/20/2011] [Accepted: 02/08/2011] [Indexed: 01/05/2023]
Abstract
Alcohol exposure in utero is a common cause of mental retardation, but the targets and mechanisms of action are poorly understood. Several lines of data point toward alterations in cortical connectivity, suggesting that axon guidance may be vulnerable to alcohol exposure. To test this, we asked whether ethanol directly affects cortical axonal growth cone responses to guidance cues. We find that even low concentrations of ethanol (12.5 mM; 57.2 mg/dl) commonly observed in social drinking prevent growth cone responses to three mechanistically independent guidance cues, Semaphorin3A, Lysophosphatidic Acid, and Netrin-1. However, this effect is highly dependent on substrate; axonal growth cones extending on an L1 cell adhesion molecule (L1CAM) substrate retain responsiveness to cues following exposure to ethanol, while those growing on poly-L-lysine or N-cadherin do not. The effects of ethanol on axon extension are, by contrast, quite modest. Quantitative assessments of the effects of ethanol on the surface distribution of L1CAM in growth cones suggest that L1CAM homophilic interactions may be particularly relevant for retaining growth cone responsiveness following ethanol exposure. Together, our findings indicate that ethanol can directly and generally alter growth cone responses to guidance cues, that a substrate of L1CAM effectively antagonizes this effect, and that cortical axonal growth cone vulnerability to ethanol may be predicted in part based on the environment through which they are extending.
Collapse
Affiliation(s)
- Bryan Sepulveda
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Ioana Carcea
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Becky Zhao
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Stephen R.J. Salton
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
- Brookdale Department of Geriatrics and Palliative Medicine, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Deanna L. Benson
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| |
Collapse
|
30
|
Twery EN, Raper JA. SDF1-induced antagonism of axonal repulsion requires multiple G-protein coupled signaling components that work in parallel. PLoS One 2011; 6:e18896. [PMID: 21556147 PMCID: PMC3083402 DOI: 10.1371/journal.pone.0018896] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 03/24/2011] [Indexed: 11/18/2022] Open
Abstract
SDF1 reduces the responsiveness of axonal growth cones to repellent guidance cues in a pertussis-toxin-sensitive, cAMP-dependent manner. Here, we show that SDF1's antirepellent effect can be blocked in embryonic chick dorsal root ganglia (DRGs) by expression of peptides or proteins inhibiting either Gαi, Gαq, or Gβγ. SDF1 antirepellent activity is also blocked by pharmacological inhibition of PLC, a common effector protein for Gαq. We also show that SDF1 antirepellent activity can be mimicked by overexpression of constitutively active Gαi, Gαq, or Gαs. These results suggest a model in which multiple G protein components cooperate to produce the cAMP levels required for SDF1 antirepellent activity.
Collapse
Affiliation(s)
- E. Naomi Twery
- Neuroscience Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jonathan A. Raper
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
31
|
Petros TJ, Bryson JB, Mason C. Ephrin-B2 elicits differential growth cone collapse and axon retraction in retinal ganglion cells from distinct retinal regions. Dev Neurobiol 2010; 70:781-94. [PMID: 20629048 DOI: 10.1002/dneu.20821] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The circuit for binocular vision and stereopsis is established at the optic chiasm, where retinal ganglion cell (RGC) axons diverge into the ipsilateral and contralateral optic tracts. In the mouse retina, ventrotemporal (VT) RGCs express the guidance receptor EphB1, which interacts with the repulsive guidance cue ephrin-B2 on radial glia at the optic chiasm to direct VT RGC axons ipsilaterally. RGCs in the ventral retina also express EphB2, which interacts with ephrin-B2, whereas dorsal RGCs express low levels of EphB receptors. To investigate how growth cones of RGCs from different retinal regions respond upon initial contact with ephrin-B2, we utilized time-lapse imaging to characterize the effects of ephrin-B2 on growth cone collapse and axon retraction in real time. We demonstrate that bath application of ephrin-B2 induces rapid and sustained growth cone collapse and axon retraction in VT RGC axons, whereas contralaterally-projecting dorsotemporal RGCs display moderate growth cone collapse and little axon retraction. Dose response curves reveal that contralaterally-projecting ventronasal axons are less sensitive to ephrin-B2 treatment compared to VT axons. Additionally, we uncovered a specific role for Rho kinase signaling in the retraction of VT RGC axons but not in growth cone collapse. The detailed characterization of growth cone behavior in this study comprises an assay for the study of Eph signaling in RGCs, and provides insight into the phenomena of growth cone collapse and axon retraction in general.
Collapse
Affiliation(s)
- Timothy J Petros
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | |
Collapse
|
32
|
Parrinello S, Napoli I, Ribeiro S, Wingfield Digby P, Fedorova M, Parkinson DB, Doddrell RDS, Nakayama M, Adams RH, Lloyd AC. EphB signaling directs peripheral nerve regeneration through Sox2-dependent Schwann cell sorting. Cell 2010; 143:145-55. [PMID: 20869108 DOI: 10.1016/j.cell.2010.08.039] [Citation(s) in RCA: 400] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 07/15/2010] [Accepted: 08/09/2010] [Indexed: 01/06/2023]
Abstract
The peripheral nervous system has astonishing regenerative capabilities in that cut nerves are able to reconnect and re-establish their function. Schwann cells are important players in this process, during which they dedifferentiate to a progenitor/stem cell and promote axonal regrowth. Here, we report that fibroblasts also play a key role. Upon nerve cut, ephrin-B/EphB2 signaling between fibroblasts and Schwann cells results in cell sorting, followed by directional collective cell migration of Schwann cells out of the nerve stumps to guide regrowing axons across the wound. Mechanistically, we find that cell-sorting downstream of EphB2 is mediated by the stemness factor Sox2 through N-cadherin relocalization to Schwann cell-cell contacts. In vivo, loss of EphB2 signaling impaired organized migration of Schwann cells, resulting in misdirected axonal regrowth. Our results identify a link between Ephs and Sox proteins, providing a mechanism by which progenitor cells can translate environmental cues to orchestrate the formation of new tissue.
Collapse
Affiliation(s)
- Simona Parrinello
- MRC Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Correction of aberrant axon growth in the developing mouse olfactory bulb. Mol Cell Neurosci 2010; 46:282-95. [PMID: 20888913 DOI: 10.1016/j.mcn.2010.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 09/23/2010] [Accepted: 09/24/2010] [Indexed: 11/23/2022] Open
Abstract
During development of the primary olfactory system, sensory axons project from the nasal cavity to the glomerular layer of the olfactory bulb. In the process axons can branch inappropriately into several glomeruli and sometimes over-shoot the glomerular layer, entering the deeper external plexiform layer. However in the adult, axons are rarely observed within the external plexiform layer. While chemorepulsive cues are proposed to restrict axons to the glomerular layer in the embryonic animal, these cues are clearly insufficient for all axons in the postnatal animal. We hypothesised that the external plexiform layer is initially an environment in which axons are able to grow but becomes increasingly inhibitory to axon growth in later postnatal development. We have determined that rather than having short localised trajectories as previously assumed, many axons that enter the external plexiform layer had considerable trajectories and projected preferentially along the ventro-dorsal and rostro-caudal axes for up to 950 μm. With increasing age, fewer axons were detected within the external plexiform layer but axons continued to be present until P17. Thus the external plexiform layer is initially an environment in which axons can extensively grow. We next tested whether the external plexiform layer became increasingly inhibitory to axon growth by microdissecting various layers of the olfactory bulb and preparing protein extracts. When assayed using olfactory epithelium explants of the same embryonic age, primary olfactory axons became increasingly inhibited by extract prepared from the external plexiform layer of increasingly older animals. These results demonstrate that primary olfactory axons can initially grow extensively in the external plexiform layer, but that during postnatal development inhibitory cues are upregulated that reduce axon growth within the external plexiform layer.
Collapse
|
34
|
A forward genetic screen in mice identifies Sema3A(K108N), which binds to neuropilin-1 but cannot signal. J Neurosci 2010; 30:5767-75. [PMID: 20410128 DOI: 10.1523/jneurosci.5061-09.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have performed a three-generation, forward genetic screen to identify recessive mutations that affect the patterning of the peripheral nervous system. Using this assay, we identified Sema3A(K108N), a novel loss-of-function allele of Sema3A. Class 3 semaphorins, which include Sema3A, are structurally conserved secreted proteins that play critical roles in the development and function of the nervous system. Sema3A(K108N) mutant mice phenocopy Sema3A-null mice, and Sema3A(K108N) protein fails to repel or collapse DRG axons in vitro. K108 is conserved among semaphorins, yet the loss-of-function effects associated with K108N are not the result of impaired expression, secretion, or binding of Sema3A to its high-affinity receptor Neuropilin-1 (Npn-1). Using in silico modeling and mutagenesis of other semaphorin family members, we predict that Sema3A(K108N) interacts poorly with the Npn-1/PlexA holoreceptor and, thus, interferes with its ability to signal at the growth cone. Therefore, through the use of a forward-genetic screen we have identified a novel allele of Sema3A that provides structural insight into the mechanism of Sema3A/Npn-1/PlexinA signaling.
Collapse
|
35
|
Joset A, Dodd DA, Halegoua S, Schwab ME. Pincher-generated Nogo-A endosomes mediate growth cone collapse and retrograde signaling. ACTA ACUST UNITED AC 2010; 188:271-85. [PMID: 20083601 PMCID: PMC2812518 DOI: 10.1083/jcb.200906089] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RhoA is activated from internalized Nogo-A to promote growth cone collapse and inhibit neurite outgrowth. Nogo-A is one of the most potent myelin-associated inhibitors for axonal growth, regeneration, and plasticity in the adult central nervous system. The Nogo-A–specific fragment NogoΔ20 induces growth cone collapse, and inhibits neurite outgrowth and cell spreading by activating RhoA. Here, we show that NogoΔ20 is internalized into neuronal cells by a Pincher- and rac-dependent, but clathrin- and dynamin-independent, mechanism. Pincher-mediated macroendocytosis results in the formation of NogoΔ20-containing signalosomes that direct RhoA activation and growth cone collapse. In compartmentalized chamber cultures, NogoΔ20 is endocytosed into neurites and retrogradely transported to the cell bodies of dorsal root ganglion neurons, triggering RhoA activation en route and decreasing phosphorylated cAMP response element binding levels in cell bodies. Thus, Pincher-dependent macroendocytosis leads to the formation of Nogo-A signaling endosomes, which act both within growth cones and after retrograde transport in the cell body to negatively regulate the neuronal growth program.
Collapse
Affiliation(s)
- Armela Joset
- Brain Research Institute, University of Zurich, Zurich, Switzerland.
| | | | | | | |
Collapse
|
36
|
Brown JA, Bridgman PC. Disruption of the cytoskeleton during Semaphorin 3A induced growth cone collapse correlates with differences in actin organization and associated binding proteins. Dev Neurobiol 2009; 69:633-46. [PMID: 19513995 DOI: 10.1002/dneu.20732] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Repulsive guidance cues induce growth cone collapse or collapse and retraction. Collapse results from disruption and loss of the actin cytoskeleton. Actin-rich regions of growth cones contain binding proteins that influence filament organization, such as Arp2/3, cortactin, and fascin, but little is known about the role that these proteins play in collapse. Here, we show that Semaphorin 3A (Sema 3A), which is repulsive to mouse dorsal root ganglion neurons, has unequal effects on actin binding proteins and their associated filaments. The immunofluorescence staining intensity of Arp-2 and cortactin decreases relative to total protein; whereas in unextracted growth cones fascin increases. Fascin and myosin IIB staining redistribute and show increased overlap. The degree of actin filament loss during collapse correlates with filament superstructures detected by rotary shadow electron microscopy. Collapse results in the loss of branched f-actin meshworks, while actin bundles are partially retained to varying degrees. Taken together with the known affects of Sema 3A on actin, this suggests a model for collapse that follows a sequence; depolymerization of actin meshworks followed by partial depolymerization of fascin associated actin bundles and their movement to the neurite to complete collapse. The relocated fascin associated actin bundles may provide the substrate for actomyosin contractions that produce retraction.
Collapse
Affiliation(s)
- Jacquelyn A Brown
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
37
|
Bush JO, Soriano P. Ephrin-B1 regulates axon guidance by reverse signaling through a PDZ-dependent mechanism. Genes Dev 2009; 23:1586-99. [PMID: 19515977 DOI: 10.1101/gad.1807209] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mutations in the ephrin-B1 gene result in craniofrontonasal syndrome (CFNS) in humans, a congenital disorder that includes a wide range of craniofacial, skeletal, and neurological malformations. In addition to the ability of ephrin-B1 to forward signal through its cognate EphB tyrosine kinase receptors, ephrin-B1 can also act as a receptor and transduce a reverse signal by either PDZ-dependent or phosphorylation-dependent mechanisms. To investigate how ephrin-B1 acts to influence development and congenital disease, we generated mice harboring a series of targeted point mutations in the ephrin-B1 gene that independently ablate specific reverse signaling pathways, while maintaining forward signaling capacity. We demonstrate that both PDZ and phosphorylation-dependent reverse signaling by ephrin-B1 are dispensable for craniofacial and skeletal development, whereas PDZ-dependent reverse signaling by ephrin-B1 is critical for the formation of a major commissural axon tract, the corpus callosum. Ephrin-B1 is strongly expressed within axons of the corpus callosum, and reverse signaling acts autonomously in cortical axons to mediate an avoidance response to its signaling partner EphB2. These results demonstrate the importance of PDZ-dependent reverse signaling for a subset of Ephrin-B1 developmental roles in vivo.
Collapse
Affiliation(s)
- Jeffrey O Bush
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | |
Collapse
|
38
|
Mintz CD, Carcea I, McNickle DG, Dickson TC, Ge Y, Salton SRJ, Benson DL. ERM proteins regulate growth cone responses to Sema3A. J Comp Neurol 2008; 510:351-66. [PMID: 18651636 DOI: 10.1002/cne.21799] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Axonal growth cones initiate and sustain directed growth in response to cues in their environment. A variety of events such as receptor internalization, kinase activation, and actin rearrangement can be stimulated by guidance cues and are essential for mediating targeted growth cone behavior. Surprisingly little is known about how such disparate actions are coordinated. Our data suggest that ezrin, radixin, and moesin (ERMs), a family of highly homologous, multifunctional proteins may be able to coordinate growth cone responses to the guidance cue Semaphorin 3A (Sema3A). We show that active ERMs concentrate asymmetrically in neocortical growth cones, are rapidly and transiently inactivated by Sema3A, and are required for Sema3A-mediated growth cone collapse and guidance. The FERM domain of active ERMs regulates internalization of the Sema3A receptor, Npn1, and its coreceptor, L1CAM, while the ERM C-terminal domain binds and caps F-actin. Our data support a model in which ERMs can coordinate membrane and actin dynamics in response to Sema3A.
Collapse
Affiliation(s)
- C David Mintz
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|