1
|
Xiong LL, Sun YF, Niu RZ, Xue LL, Chen L, Huangfu LR, Li J, Wang YY, Liu X, Wang WY, Zuo ZF, Wang TH. Cellular Characterization and Interspecies Evolution of the Tree Shrew Retina across Postnatal Lifespan. RESEARCH (WASHINGTON, D.C.) 2024; 7:0536. [PMID: 39574940 PMCID: PMC11579486 DOI: 10.34133/research.0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/20/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024]
Abstract
Tree shrews (TSs) possess a highly developed visual system. Here, we establish an age-related single-cell RNA sequencing atlas of retina cells from 15 TSs, covering 6 major retina cell classes and 3 glial cell types. An age effect is observed on the cell subset composition and gene expression pattern. We then verify the cell subtypes and identify specific markers in the TS retina including CA10 for bipolar cells, MEGF11 for H1 horizontal cells, and SLIT2, RUNX1, FOXP2, and SPP1 for retinal ganglion cell subpopulations. The cross-species analysis elucidates the cell type-specific transcriptional programs, different cell compositions, and cell communications. The comparisons also reveal that TS cones and subclasses of bipolar and amacrine cells exhibit the closest relationship with humans and macaques. Our results suggests that TS could be used as a better disease model to understand age-dependent cellular and genetic mechanisms of the retina, particularly for the retinal diseases associated with cones.
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Department of Anesthesiology, Research Institute of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Anesthesiology,
The Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Yi-Fei Sun
- Department of Urology,
the Second Affiliated Hospital of Kunming Medical University, Kunming 650500, China
| | - Rui-Ze Niu
- Mental Health Center of Kunming Medical University, Kunming 650034, Yunnan, China
| | - Lu-Lu Xue
- State Key Lab of Biotherapy, West China Hospital,
Sichuan University, Chengdu 610041, Sichuan, China
| | - Li Chen
- Department of Anesthesiology, Research Institute of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Li-Ren Huangfu
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Jing Li
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Yu-Ying Wang
- Department of Anatomy, College of Basic Medicine, Jinzhou Medical University, Jinzhou 121001, Liaoning, China
| | - Xin Liu
- Department of Anatomy, College of Basic Medicine, Jinzhou Medical University, Jinzhou 121001, Liaoning, China
| | - Wen-Yuan Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Science, Shanghai 200032, China
| | - Zhong-Fu Zuo
- Department of Anatomy, College of Basic Medicine, Jinzhou Medical University, Jinzhou 121001, Liaoning, China
| | - Ting-Hua Wang
- Department of Anesthesiology, Research Institute of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
- Department of Anatomy, College of Basic Medicine, Jinzhou Medical University, Jinzhou 121001, Liaoning, China
| |
Collapse
|
2
|
Galindo-Torres P, Rosas C, Ramos-Rodríguez S, Galindo-Sánchez CE. Chronic thermal stress on Octopus maya embryos down-regulates epigenome-related genes and those involved in the nervous system development and morphogenesis. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101332. [PMID: 39366120 DOI: 10.1016/j.cbd.2024.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/06/2024]
Abstract
Red Octopus maya is strongly influenced by temperature. Recent studies have reported negative reproduction effects on males and females when exposed to temperatures higher than 27 °C. Embryos under thermal stress show morphological and physiological alterations; similar phenotypes have been reported in embryos from stressed females, evidencing transgenerational consequences. Transcriptomic profiles were characterized along embryo development during normal-under thermal stress and epigenetic alterations through DNA methylation and damage quantification. Total RNA in organogenesis, activation, and growth stages in control and thermal stress were sequenced with Illumina RNA-Seq. Similarly, total DNA was used for DNA methylation and damage quantification between temperatures and embryo stages. Differential gene expression analyses showed that embryos express genes associated with oxygen transport, morphogenesis, nervous system, neuroendocrine cell differentiation, spermatogenesis, and male sex differentiation. Conversely, embryos turn off genes involved mainly in nervous system development, morphogenesis, and gene expression regulation when exposed to thermal stress - consistent with O. maya embryo phenotypes showing abnormal arms, eyes, and body development. No significant differences were observed in quantifying DNA methylation between temperatures but they were for DNA damage quantification. Epigenetic alterations are hypothesized to occur since several genes found downregulated belong to the epigenetic machinery but at histone tail level.
Collapse
Affiliation(s)
- Pavel Galindo-Torres
- Centro de Investigacion Cientifica y de Educacion Superior de Ensenada (CICESE), Carretera Tijuana-Ensenada No. 3918, Zona Playitas, Ensenada, Baja California CP 22860, Mexico.
| | - Carlos Rosas
- Unidad Multidisciplinaria de Docencia e Investigacion (UMDI), Facultad de Ciencias, Universidad Nacional Autonoma de Mexico (UNAM), Puerto DE Abrigo s/n, Sisal, Hunucma, Yucatan CP97355, Mexico.
| | - Sadot Ramos-Rodríguez
- Centro de Investigacion Cientifica y de Educacion Superior de Ensenada (CICESE), Carretera Tijuana-Ensenada No. 3918, Zona Playitas, Ensenada, Baja California CP 22860, Mexico.
| | - Clara E Galindo-Sánchez
- Centro de Investigacion Cientifica y de Educacion Superior de Ensenada (CICESE), Carretera Tijuana-Ensenada No. 3918, Zona Playitas, Ensenada, Baja California CP 22860, Mexico.
| |
Collapse
|
3
|
Le VH, Orniacki C, Murcia-Belmonte V, Denti L, Schütz D, Stumm R, Ruhrberg C, Erskine L. CXCL12 promotes the crossing of retinal ganglion cell axons at the optic chiasm. Development 2024; 151:dev202446. [PMID: 38095299 PMCID: PMC10820821 DOI: 10.1242/dev.202446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/07/2023] [Indexed: 01/13/2024]
Abstract
Binocular vision requires the segregation of retinal ganglion cell (RGC) axons extending from the retina into the ipsilateral and contralateral optic tracts. RGC axon segregation occurs at the optic chiasm, which forms at the ventral diencephalon midline. Using expression analyses, retinal explants and genetically modified mice, we demonstrate that CXCL12 (SDF1) is required for axon segregation at the optic chiasm. CXCL12 is expressed by the meninges bordering the optic pathway, and CXCR4 by both ipsilaterally and contralaterally projecting RGCs. CXCL12 or ventral diencephalon meninges potently promoted axon outgrowth from both ipsilaterally and contralaterally projecting RGCs. Further, a higher proportion of axons projected ipsilaterally in mice lacking CXCL12 or its receptor CXCR4 compared with wild-type mice as a result of misrouting of presumptive contralaterally specified RGC axons. Although RGCs also expressed the alternative CXCL12 receptor ACKR3, the optic chiasm developed normally in mice lacking ACKR3. Our data support a model whereby meningeal-derived CXCL12 helps drive axon growth from CXCR4-expressing RGCs towards the diencephalon midline, enabling contralateral axon growth. These findings further our understanding of the molecular and cellular mechanisms controlling optic pathway development.
Collapse
Affiliation(s)
- Viet-Hang Le
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| | - Clarisse Orniacki
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| | - Verónica Murcia-Belmonte
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Dagmar Schütz
- Institute for Pharmacology/Toxicology, Jena University Hospital,Drackendorfer Str. 1, D-07747 Jena, Germany
| | - Ralf Stumm
- Institute for Pharmacology/Toxicology, Jena University Hospital,Drackendorfer Str. 1, D-07747 Jena, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Lynda Erskine
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| |
Collapse
|
4
|
Paşcalău R, Badea TC. Signaling - transcription interactions in mouse retinal ganglion cells early axon pathfinding -a literature review. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1180142. [PMID: 38983012 PMCID: PMC11182120 DOI: 10.3389/fopht.2023.1180142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/21/2023] [Indexed: 07/11/2024]
Abstract
Sending an axon out of the eye and into the target brain nuclei is the defining feature of retinal ganglion cells (RGCs). The literature on RGC axon pathfinding is vast, but it focuses mostly on decision making events such as midline crossing at the optic chiasm or retinotopic mapping at the target nuclei. In comparison, the exit of RGC axons out of the eye is much less explored. The first checkpoint on the RGC axons' path is the optic cup - optic stalk junction (OC-OS). OC-OS development and the exit of the RGC pioneer axons out of the eye are coordinated spatially and temporally. By the time the optic nerve head domain is specified, the optic fissure margins are in contact and the fusion process is ongoing, the first RGCs are born in its proximity and send pioneer axons in the optic stalk. RGC differentiation continues in centrifugal waves. Later born RGC axons fasciculate with the more mature axons. Growth cones at the end of the axons respond to guidance cues to adopt a centripetal direction, maintain nerve fiber layer restriction and to leave the optic cup. Although there is extensive information on OC-OS development, we still have important unanswered questions regarding its contribution to the exit of the RGC axons out of the eye. We are still to distinguish the morphogens of the OC-OS from the axon guidance molecules which are expressed in the same place at the same time. The early RGC transcription programs responsible for axon emergence and pathfinding are also unknown. This review summarizes the molecular mechanisms for early RGC axon guidance by contextualizing mouse knock-out studies on OC-OS development with the recent transcriptomic studies on developing RGCs in an attempt to contribute to the understanding of human optic nerve developmental anomalies. The published data summarized here suggests that the developing optic nerve head provides a physical channel (the closing optic fissure) as well as molecular guidance cues for the pioneer RGC axons to exit the eye.
Collapse
Affiliation(s)
- Raluca Paşcalău
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- Ophthalmology Clinic, Cluj County Emergency Hospital, Cluj-Napoca, Romania
| | - Tudor Constantin Badea
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- National Center for Brain Research, Institutul de Cercetări pentru Inteligență Artificială, Romanian Academy, Bucharest, Romania
| |
Collapse
|
5
|
Kwak HJ, Medina-Jiménez BI, Park SC, Kim JH, Jeong GH, Jeon MJ, Kim S, Kim JW, Weisblat DA, Cho SJ. Slit-Robo expression in the leech nervous system: insights into eyespot evolution. Cell Biosci 2023; 13:70. [PMID: 37013648 PMCID: PMC10071614 DOI: 10.1186/s13578-023-01019-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/26/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Slit and Robo are evolutionarily conserved ligand and receptor proteins, respectively, but the number of slit and robo gene paralogs varies across recent bilaterian genomes. Previous studies indicate that this ligand-receptor complex is involved in axon guidance. Given the lack of data regarding Slit/Robo in the Lophotrochozoa compared to Ecdysozoa and Deuterostomia, the present study aims to identify and characterize the expression of Slit/Robo orthologs in leech development. RESULTS We identified one slit (Hau-slit), and two robo genes (Hau-robo1 and Hau-robo2), and characterized their expression spatiotemporally during the development of the glossiphoniid leech Helobdella austinensis. Throughout segmentation and organogenesis, Hau-slit and Hau-robo1 are broadly expressed in complex and roughly complementary patterns in the ventral and dorsal midline, nerve ganglia, foregut, visceral mesoderm and/or endoderm of the crop, rectum and reproductive organs. Before yolk exhaustion, Hau-robo1 is also expressed where the pigmented eye spots will later develop, and Hau-slit is expressed in the area between these future eye spots. In contrast, Hau-robo2 expression is extremely limited, appearing first in the developing pigmented eye spots, and later in the three additional pairs of cryptic eye spots in head region that never develop pigment. Comparing the expression of robo orthologs between H. austinensis and another glossiphoniid leech, Alboglossiphonia lata allows to that robo1 and robo2 operate combinatorially to differentially specify pigmented and cryptic eyespots within the glossiphoniid leeches. CONCLUSIONS Our results support a conserved role in neurogenesis, midline formation and eye spot development for Slit/Robo in the Lophotrochozoa, and provide relevant data for evo-devo studies related to nervous system evolution.
Collapse
Affiliation(s)
- Hee-Jin Kwak
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Department of Ecology, Evolution and Behavior, Faculty of Science, Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, 9190401, Jerusalem, Israel
| | - Brenda I Medina-Jiménez
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Department of Earth Sciences, Paleobiology, Geocentrum, Uppsala University, Villavägen 16, 75236, Uppsala, Sweden
| | - Soon Cheol Park
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Hyeuk Kim
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Wildlife Disease Response Team, National Institute of Wildlife Disease Control and Prevention, Incheon, 22689, Republic of Korea
| | - Geon-Hwi Jeong
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Mi-Jeong Jeon
- National Institute of Biological Resources, Environmental Research Complex, Incheon, 22689, Republic of Korea
| | - Sangil Kim
- Museum of Comparative Zoology and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Jung-Woong Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - David A Weisblat
- Department of Molecular and Cell Biology, University of California, 385 Weill Hall, Berkeley, CA, 94720-3200, USA.
| | - Sung-Jin Cho
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
6
|
Subramani M, Hook MV, Rajamoorthy M, Qiu F, Ahmad I. Human Retinal Ganglion Cells Respond to Evolutionarily Conserved Chemotropic Cues for Intra Retinal Guidance and Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526677. [PMID: 36778442 PMCID: PMC9915675 DOI: 10.1101/2023.02.01.526677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Retinal ganglion cells (RGCs) connect the retina with the higher centers in the brain for visual perception. Their degeneration leads to irreversible vision loss in glaucoma patients. Since human RGCs (hRGCs) are born during fetal development and connections with the central targets are established before birth, the mechanism underlying their axon growth and guidance remains poorly understood. Here, using RGCs directly generated from human embryonic stem cells, we demonstrate that hRGCs express a battery of guidance receptors. These receptors allow hRGCs to read the spatially arrayed chemotropic cues in the developing rat retina for the centripetal orientation of axons toward the optic disc, suggesting that the mechanism of intra-retinal guidance is conserved in hRGCs. The centripetal orientation of hRGCs axons is not only in response to chemo-repulsion but also involves chemo-attraction, mediated by Netrin-1/DCC interactions. The spatially arrayed chemotropic cues differentially influence hRGCs physiological responses, suggesting that neural activity of hRGCs may facilitate axon growth during inter-retinal guidance. Additionally, we demonstrate that Netrin-1/DCC interactions, besides promoting axon growth, facilitate hRGCs axon regeneration by recruiting the mTOR signaling pathway. The diverse influence of Netrin-1/DCC interactions ranging from axon growth to regeneration may involve recruitment of multiple intracellular signaling pathways as revealed by transcriptome analysis of hRGCs. From the perspective of ex-vivo stem cell approach to glaucomatous degeneration, our findings posit that ex-vivo generated human RGCs are capable of reading the intra-retinal cues for guidance toward the optic disc, the first step toward connecting with the central target to restore vision.
Collapse
|
7
|
Schwend T. Wiring the ocular surface: A focus on the comparative anatomy and molecular regulation of sensory innervation of the cornea. Differentiation 2023:S0301-4681(23)00010-5. [PMID: 36997455 DOI: 10.1016/j.diff.2023.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
The cornea is richly innervated with sensory nerves that function to detect and clear harmful debris from the surface of the eye, promote growth and survival of the corneal epithelium and hasten wound healing following ocular disease or trauma. Given their importance to eye health, the neuroanatomy of the cornea has for many years been a source of intense investigation. Resultantly, complete nerve architecture maps exist for adult human and many animal models and these maps reveal few major differences across species. Interestingly, recent work has revealed considerable variation across species in how sensory nerves are acquired during developmental innervation of the cornea. Highlighting such species-distinct key differences, but also similarities, this review provides a full, comparative anatomy analysis of sensory innervation of the cornea for all species studied to date. Further, this article comprehensively describes the molecules that have been shown to guide and direct nerves toward, into and through developing corneal tissue as the final architectural pattern of the cornea's neuroanatomy is established. Such knowledge is useful for researchers and clinicians seeking to better understand the anatomical and molecular basis of corneal nerve pathologies and to hasten neuro-regeneration following infection, trauma or surgery that damage the ocular surface and its corneal nerves.
Collapse
|
8
|
Zebrafish Slit2 and Slit3 Act Together to Regulate Retinal Axon Crossing at the Midline. J Dev Biol 2022; 10:jdb10040041. [PMID: 36278546 PMCID: PMC9590056 DOI: 10.3390/jdb10040041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Slit-Robo signaling regulates midline crossing of commissural axons in different systems. In zebrafish, all retinofugal axons cross at the optic chiasm to innervate the contralateral tectum. Here, the mutant for the Robo2 receptor presents severe axon guidance defects, which were not completely reproduced in a Slit2 ligand null mutant. Since slit3 is also expressed around this area at the stage of axon crossing, we decided to analyze the possibility that it collaborates with Slit2 in this process. We found that the disruption of slit3 expression by sgRNA-Cas9 injection caused similar, albeit slightly milder, defects than those of the slit2 mutant, while the same treatment in the slit2−/−mz background caused much more severe defects, comparable to those observed in robo2 mutants. Tracking analysis of in vivo time-lapse experiments indicated differential but complementary functions of these secreted factors in the correction of axon turn errors around the optic chiasm. Interestingly, RT-qPCR analysis showed a mild increase in slit2 expression in slit3-deficient embryos, but not the opposite. Our observations support the previously proposed “repulsive channel” model for Slit-Robo action at the optic chiasm, with both Slits acting in different manners, most probably relating to their different spatial expression patterns.
Collapse
|
9
|
Rafipay A, Dun X, Parkinson DB, Erskine L, Vargesson N. Knockdown of slit signaling during limb development leads to a reduction in humerus length. Dev Dyn 2021; 250:1340-1357. [DOI: 10.1002/dvdy.284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Alexandra Rafipay
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| | - Xin‐Peng Dun
- Peninsula Medical School, Faculty of Health University of Plymouth Plymouth UK
| | - David B Parkinson
- Peninsula Medical School, Faculty of Health University of Plymouth Plymouth UK
| | - Lynda Erskine
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| | - Neil Vargesson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| |
Collapse
|
10
|
Conceição R, Evans RS, Pearson CS, Hänzi B, Osborne A, Deshpande SS, Martin KR, Barber AC. Expression of Developmentally Important Axon Guidance Cues in the Adult Optic Chiasm. Invest Ophthalmol Vis Sci 2019; 60:4727-4739. [PMID: 31731293 PMCID: PMC6859889 DOI: 10.1167/iovs.19-26732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Regeneration of optic nerve axons after injury can be facilitated by several approaches, but misguidance at the optic chiasm is often observed. We characterized guidance cues in the embryonic visual system and adult optic chiasm before and after optic nerve crush (ONC) injury to better understand barriers to optic nerve regeneration in adults. Methods Radial glial (RC2/BLBP/Slit1), developmental (Pax2) and extracellular markers (CSPG: H2B/CS-56) were assessed in C57BL/6J mice by immunohistochemistry. RC2, BLBP, Slit1, and CSPG are known inhibitory guidance cues while Pax2 is a permissive guidance cue. Results At embryonic day 15.5 (E.15.5), RC2 and BLBP were identified superior to, and extending through, the optic chiasm. The optic chiasm was BLBP-ve in adult uninjured mice but BLBP+ve in adult mice 10 days after ONC injury. The reverse was true for RC2. Both BLBP and RC2 were absent in adult mice 6 weeks post-ONC. Slit1 was present in the optic chiasm midline and optic tracts in embryonic samples but was absent in uninjured adult tissue. Slit1 was observed superior to and at the midline of the optic chiasm 10 days post-ONC but absent 6 weeks after injury. Pax2 was expressed at the junction between the optic nerve and optic chiasm in embryonic brain tissue. In embryonic sections, CS-56 was observed at the junction between the optic chiasm and optic tract, and immediately superior to the optic chiasm. Both 2H6 and CS-56 staining was absent in uninjured and ONC-injured adult brains. Conclusion Differences in guidance cue expression during development, in adulthood and after injury may contribute to misguidance of regenerating RGC axons in the adult optic chiasm.
Collapse
Affiliation(s)
- Raquel Conceição
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Rachel S Evans
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Craig S Pearson
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom.,Laboratory of Developmental Neurobiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Barbara Hänzi
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Andrew Osborne
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Sarita S Deshpande
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Keith R Martin
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom.,Centre for Eye Research Australia, Melbourne, Australia.,University of Melbourne, Melbourne, Australia.,Department of Ophthalmology, NIHR Biomedical Research Centre and Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, United Kingdom
| | - Amanda C Barber
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| |
Collapse
|
11
|
Herrera E, Agudo-Barriuso M, Murcia-Belmonte V. Cranial Pair II: The Optic Nerves. Anat Rec (Hoboken) 2018; 302:428-445. [DOI: 10.1002/ar.23922] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/19/2017] [Accepted: 05/14/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina; Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca); Murcia Spain
| | - Verónica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| |
Collapse
|
12
|
Diao Y, Chen Y, Zhang P, Cui L, Zhang J. Molecular guidance cues in the development of visual pathway. Protein Cell 2017; 9:909-929. [PMID: 29181831 PMCID: PMC6208478 DOI: 10.1007/s13238-017-0490-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/30/2017] [Indexed: 01/23/2023] Open
Abstract
70%–80% of our sensory input comes from vision. Light hit the retina at the back of our eyes and the visual information is relayed into the dorsal lateral geniculate nuclei (dLGN) and primary visual cortex (V1) thereafter, constituting the image-forming visual circuit. Molecular cues are one of the key factors to guide the wiring and refinement of the image-forming visual circuit during pre- and post-embryonic stages. Distinct molecular cues are involved in different developmental stages and nucleus, suggesting diverse guidance mechanisms. In this review, we summarize molecular guidance cues throughout the image-forming visual circuit, including chiasm determination, eye-specific segregation and refinement in the dLGN, and at last the reciprocal connections between the dLGN and V1.
Collapse
Affiliation(s)
- Yupu Diao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yuqing Chen
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Peijun Zhang
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Liyuan Cui
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jiayi Zhang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Guidance of retinal axons in mammals. Semin Cell Dev Biol 2017; 85:48-59. [PMID: 29174916 DOI: 10.1016/j.semcdb.2017.11.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 11/21/2022]
Abstract
In order to navigate through the surrounding environment many mammals, including humans, primarily rely on vision. The eye, composed of the choroid, sclera, retinal pigmented epithelium, cornea, lens, iris and retina, is the structure that receives the light and converts it into electrical impulses. The retina contains six major types of neurons involving in receiving and modifying visual information and passing it onto higher visual processing centres in the brain. Visual information is relayed to the brain via the axons of retinal ganglion cells (RGCs), a projection known as the optic pathway. The proper formation of this pathway during development is essential for normal vision in the adult individual. Along this pathway there are several points where visual axons face 'choices' in their direction of growth. Understanding how these choices are made has advanced significantly our knowledge of axon guidance mechanisms. Thus, the development of the visual pathway has served as an extremely useful model to reveal general principles of axon pathfinding throughout the nervous system. However, due to its particularities, some cellular and molecular mechanisms are specific for the visual circuit. Here we review both general and specific mechanisms involved in the guidance of mammalian RGC axons when they are traveling from the retina to the brain to establish precise and stereotyped connections that will sustain vision.
Collapse
|
14
|
Lee KH, Gee HY, Shin JI. Genetics of vesicoureteral reflux and congenital anomalies of the kidney and urinary tract. Investig Clin Urol 2017; 58:S4-S13. [PMID: 28612055 PMCID: PMC5468264 DOI: 10.4111/icu.2017.58.s1.s4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/20/2017] [Indexed: 01/17/2023] Open
Abstract
The definition of congenital anomalies of the kidney and urinary tract (CAKUT) is the disease of structural malformations in the kidney and/or urinary tract containing vesicoureteral reflux (VUR). These anomalies can cause pediatric chronic kidney disease. However, the pathogenesis of CAKUT is not well understood, because identifying the genetic architecture of CAKUT is difficult due to the phenotypic heterogeneity and multifactorial genetic penetrance. We describe the current genetic basis and mechanisms of CAKUT including VUR via approaching the steps and signaling pathways of kidney developmental processes. We also focus on the newly developed strategies and challenges to fully address the role of the associated genes in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Keum Hwa Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea.,Department of Pediatric Nephrology, Severance Children's Hospital, Seoul, Korea.,Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Korea
| | - Heon Yung Gee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea.,Department of Pediatric Nephrology, Severance Children's Hospital, Seoul, Korea.,Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Ke C, Gao F, Tian X, Li C, Shi D, He W, Tian Y. Slit2/Robo1 Mediation of Synaptic Plasticity Contributes to Bone Cancer Pain. Mol Neurobiol 2017; 54:295-307. [PMID: 26738857 DOI: 10.1007/s12035-015-9564-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/29/2015] [Indexed: 12/11/2022]
Abstract
Synaptic plasticity is fundamental to spinal sensitivity of bone cancer pain. Here, we have shown that excitatory synaptogenesis contributes to bone cancer pain. New synapse formation requires neurite outgrowth and an interaction between axons and dendrites, accompanied by the appositional organization of presynaptic and postsynaptic specializations. We have shown that Slit2, Robo1, and RhoA act as such cues that promote neurite outgrowth and guide the axon for synapse formation. Sarcoma inoculation induces excitatory synaptogenesis and bone cancer pain which are reversed by Slit2 knockdown but aggravated by Robo1 knockdown. Synaptogenesis of cultured neurons are inhibited by Slit2 knockdown but enhanced by Robo1 knockdown. Sarcoma implantation induces an increase in Slit2 and decreases Robo1 and RhoA, while Slit2 knockdown results in an increase of Robo1 and RhoA. These results have demonstrated a molecular mechanism of synaptogenesis in bone cancer pain.
Collapse
Affiliation(s)
- Changbin Ke
- Institute of Anesthesiology and Pain (IAP) and Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan City, 442000, Hubei Province, China
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Caijuan Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dai Shi
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wensheng He
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuke Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
16
|
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
|
17
|
McConnell RE, Edward van Veen J, Vidaki M, Kwiatkowski AV, Meyer AS, Gertler FB. A requirement for filopodia extension toward Slit during Robo-mediated axon repulsion. J Cell Biol 2016; 213:261-74. [PMID: 27091449 PMCID: PMC5084274 DOI: 10.1083/jcb.201509062] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 03/04/2016] [Indexed: 12/11/2022] Open
Abstract
Axons navigate long distances through complex 3D environments to interconnect the nervous system during development. Although the precise spatiotemporal effects of most axon guidance cues remain poorly characterized, a prevailing model posits that attractive guidance cues stimulate actin polymerization in neuronal growth cones whereas repulsive cues induce actin disassembly. Contrary to this model, we find that the repulsive guidance cue Slit stimulates the formation and elongation of actin-based filopodia from mouse dorsal root ganglion growth cones. Surprisingly, filopodia form and elongate toward sources of Slit, a response that we find is required for subsequent axonal repulsion away from Slit. Mechanistically, Slit evokes changes in filopodium dynamics by increasing direct binding of its receptor, Robo, to members of the actin-regulatory Ena/VASP family. Perturbing filopodium dynamics pharmacologically or genetically disrupts Slit-mediated repulsion and produces severe axon guidance defects in vivo. Thus, Slit locally stimulates directional filopodial extension, a process that is required for subsequent axonal repulsion downstream of the Robo receptor.
Collapse
Affiliation(s)
- Russell E McConnell
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 01239
| | - J Edward van Veen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 01239 Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 01239
| | - Marina Vidaki
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 01239
| | - Adam V Kwiatkowski
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 01239
| | - Aaron S Meyer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 01239 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 01239
| | - Frank B Gertler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 01239 Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 01239
| |
Collapse
|
18
|
Wu H, Barik A, Lu Y, Shen C, Bowman A, Li L, Sathyamurthy A, Lin TW, Xiong WC, Mei L. Slit2 as a β-catenin/Ctnnb1-dependent retrograde signal for presynaptic differentiation. eLife 2015; 4. [PMID: 26159615 PMCID: PMC4498096 DOI: 10.7554/elife.07266] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 06/18/2015] [Indexed: 12/12/2022] Open
Abstract
Neuromuscular junction formation requires proper interaction between motoneurons and muscle cells. β-Catenin (Ctnnb1) in muscle is critical for motoneuron differentiation; however, little is known about the relevant retrograde signal. In this paper, we dissected which functions of muscle Ctnnb1 are critical by an in vivo transgenic approach. We show that Ctnnb1 mutant without the transactivation domain was unable to rescue presynaptic deficits of Ctnnb1 mutation, indicating the involvement of transcription regulation. On the other hand, the cell-adhesion function of Ctnnb1 is dispensable. We screened for proteins that may serve as a Ctnnb1-directed retrograde factor and identified Slit2. Transgenic expression of Slit2 specifically in the muscle was able to diminish presynaptic deficits by Ctnnb1 mutation in mice. Slit2 immobilized on beads was able to induce synaptophysin puncta in axons of spinal cord explants. Together, these observations suggest that Slit2 serves as a factor utilized by muscle Ctnnb1 to direct presynaptic differentiation. DOI:http://dx.doi.org/10.7554/eLife.07266.001 Motor nerves are like electrical wires that connect our spinal cord to the muscles in our body. These nerves communicate with muscles across a connection called the neuromuscular junction. To first form a neuromuscular junction, the motor nerves and muscles each produce molecular cues that tell each other to do their part to build a connection. Beta-catenin in the muscle is known to regulate motor nerve development. However, beta-catenin has two different roles: it helps to coordinate whether neighboring cells stick together, and it can regulate which genes are ‘transcribed’ to produce proteins. It was not known which of these roles is necessary for forming neuromuscular junctions. Wu, Barik et al. now investigate this question by creating mice with mutant forms of beta-catenin in their muscles. Some mice had muscle beta-catenin that could not help cells stick together, and others had beta-catenin that could not control gene transcription. Only mutations that affected the ability of beta-catenin to control transcription caused abnormalities in the neuromuscular junction. However, these problems could be fixed by adding either normal beta-catenin or the mutant form that cannot help cells stick together. Wu, Barik et al. then used molecular tools to explore which genes are turned on by beta-catenin. The experiments showed that beta-catenin causes muscle fibers to produce a protein called Slit2—a developmental cue that controls where neurons grow. Furthermore, the neuromuscular junction defects found in mice without beta-catenin in their muscles could be reduced by making the muscle fibers produce more Slit2. However, not all defects in beta-catenin mutant mice are rescued by Slit2. Future research is needed to identify other beta-catenin-controlled signals and to determine whether such a pathway is altered in neuromuscular disorders. DOI:http://dx.doi.org/10.7554/eLife.07266.002
Collapse
Affiliation(s)
- Haitao Wu
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing, China
| | - Arnab Barik
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| | - Yisheng Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| | - Chengyong Shen
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| | - Andrew Bowman
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| | - Lei Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| | - Anupama Sathyamurthy
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| | - Thiri W Lin
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| | - Wen-Cheng Xiong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| | - Lin Mei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, United States
| |
Collapse
|
19
|
Zhang C, Guo H, Li B, Sui C, Zhang Y, Xia X, Qin Y, Ye L, Xie F, Wang H, Yuan M, Yuan L, Ye J. Effects of Slit3 silencing on the invasive ability of lung carcinoma A549 cells. Oncol Rep 2015; 34:952-60. [PMID: 26045181 DOI: 10.3892/or.2015.4031] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/18/2015] [Indexed: 11/05/2022] Open
Abstract
Slit proteins function as chemorepellents in axon guidance and neuronal migration by binding to cognate Robo receptors. The Slit/Robo signaling pathway is also involved in the regulation of tumor cell metastasis. However, whether the Slit/Robo signaling pathway exerts prometastatic or antimetastasis functions remains controversial. To date, most of the research on Slit/Robo has focused on Slit2, and the effects of Slit3 on metastasis remain largely unknown. Based on the Oncomine database, overall expression of Slit3 is low in tumor tissues compared to its level in normal tissues. The underlying mechanism for slit3 silencing in tumor tissues is likely related to hypermethylation of the slit3 promoter. However, lung carcinomas appear to be an exception. Several studies have reported that the frequency of Slit3 methylation in lung cancers is far lower than the frequency of Slit2. In the present study, high Slit3 expression at the mRNA level, yet not at the protein level, was detected in lung adenocarcinoma A549 cells. The function of Slit3 in tumor migration and invasion was examined by silencing of Slit3 expression in A549 cells. Silencing of Slit3 promoted proliferation, migration and invasion of A549 cells and induced epithelial-mesenchymal transition by downregulation of E-cadherin and upregulation of vimentin. The inhibitory effects of Slit3 on tumor migration and invasion are likely related to matrix metalloproteinases (MMPs). Silencing of Slit3 in the A549 cells enhanced MMP2 and MMP9 expression. These results indicate that Slit3 is a potential tumor suppressor in lung adenocarcinoma.
Collapse
Affiliation(s)
- Chao Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Hui Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Bin Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Chengzhi Sui
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, P.R. China
| | - Yuan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Xianyuan Xia
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Ying Qin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Liying Ye
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Fu'an Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Heng Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Mingjing Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Li Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| | - Jun Ye
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, P.R. China
| |
Collapse
|
20
|
Abstract
The visual system is beautifully crafted to transmit information of the external world to visual processing and cognitive centers in the brain. For visual information to be relayed to the brain, a series of axon pathfinding events must take place to ensure that the axons of retinal ganglion cells, the only neuronal cell type in the retina that sends axons out of the retina, find their way out of the eye to connect with targets in the brain. In the past few decades, the power of molecular and genetic tools, including the generation of genetically manipulated mouse lines, have multiplied our knowledge about the molecular mechanisms involved in the sculpting of the visual system. Here, we review major advances in our understanding of the mechanisms controlling the differentiation of RGCs, guidance of their axons from the retina to the primary visual centers, and the refinement processes essential for the establishment of topographic maps and eye-specific axon segregation. Human disorders, such as albinism and achiasmia, that impair RGC axon growth and guidance and, thus, the establishment of a fully functioning visual system will also be discussed.
Collapse
Affiliation(s)
- Lynda Erskine
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Scotland, UK
| | - Eloisa Herrera
- Instituto de Neurosciencias de Alicante, CSIC-UMH, San Juan de Alicante, Spain
| |
Collapse
|
21
|
Zhao H, Anand AR, Ganju RK. Slit2-Robo4 pathway modulates lipopolysaccharide-induced endothelial inflammation and its expression is dysregulated during endotoxemia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:385-93. [PMID: 24272999 PMCID: PMC3908786 DOI: 10.4049/jimmunol.1302021] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The secretory protein Slit2 and its receptors Robo1 and Robo4 are considered to regulate mobility and permeability of endothelial cells and other cell types. However, the roles of Slit2 and its two receptors in endothelial inflammatory responses remain to be clarified. In this study, we show that, in primary HUVECs, Slit2 represses LPS-induced secretion of certain inflammatory cytokines/chemokines, cell adhesion molecule ICAM-1 upregulation, and monocyte adhesion. Slit2's anti-inflammatory effect is mediated by its dominant endothelial-specific receptor Robo4. However, the minor receptor Robo1 has proinflammatory properties and is downregulated by Slit2 via targeting of miR-218. Elucidation of molecular mechanism reveals that Slit2 represses inflammatory responses by inhibiting the Pyk2-NF-κB pathway downstream of LPS-TLR4. Further studies reveal that LPS enhances endothelial inflammation by downregulating the anti-inflammatory Slit2 and Robo4 in HUVECs in vitro, as well as in arterial endothelial cells and liver in vivo during endotoxemia. These results suggest that Slit2-Robo4 signaling is important in regulating LPS-induced endothelial inflammation, and LPS, in turn, enhances inflammation by interfering with the expression of the anti-inflammatory Slit2-Robo4 during the disease state. This implies that Slit2-Robo4 is a key regulator of endothelial inflammation, and its dysregulation during endotoxemia is a novel mechanism for LPS-induced vascular pathogenesis.
Collapse
Affiliation(s)
- Helong Zhao
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | | | | |
Collapse
|
22
|
Gene expression analysis of induced pluripotent stem cells from aneuploid chromosomal syndromes. BMC Genomics 2013; 14 Suppl 5:S8. [PMID: 24564826 PMCID: PMC3852284 DOI: 10.1186/1471-2164-14-s5-s8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Human aneuploidy is the leading cause of early pregnancy loss, mental retardation, and multiple congenital anomalies. Due to the high mortality associated with aneuploidy, the pathophysiological mechanisms of aneuploidy syndrome remain largely unknown. Previous studies focused mostly on whether dosage compensation occurs, and the next generation transcriptomics sequencing technology RNA-seq is expected to eventually uncover the mechanisms of gene expression regulation and the related pathological phenotypes in human aneuploidy. Results Using next generation transcriptomics sequencing technology RNA-seq, we profiled the transcriptomes of four human aneuploid induced pluripotent stem cell (iPSC) lines generated from monosomy × (Turner syndrome), trisomy 8 (Warkany syndrome 2), trisomy 13 (Patau syndrome), and partial trisomy 11:22 (Emanuel syndrome) as well as two umbilical cord matrix iPSC lines as euploid controls to examine how phenotypic abnormalities develop with aberrant karyotype. A total of 466 M (50-bp) reads were obtained from the six iPSC lines, and over 13,000 mRNAs were identified by gene annotation. Global analysis of gene expression profiles and functional analysis of differentially expressed (DE) genes were implemented. Over 5000 DE genes are determined between aneuploidy and euploid iPSCs respectively while 9 KEGG pathways are overlapped enriched in four aneuploidy samples. Conclusions Our results demonstrate that the extra or missing chromosome has extensive effects on the whole transcriptome. Functional analysis of differentially expressed genes reveals that the genes most affected in aneuploid individuals are related to central nervous system development and tumorigenesis.
Collapse
|
23
|
James G, Foster SR, Key B, Beverdam A. The expression pattern of EVA1C, a novel Slit receptor, is consistent with an axon guidance role in the mouse nervous system. PLoS One 2013; 8:e74115. [PMID: 24040182 PMCID: PMC3767613 DOI: 10.1371/journal.pone.0074115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 08/01/2013] [Indexed: 11/18/2022] Open
Abstract
The Slit/Robo axon guidance families play a vital role in the formation of neural circuitry within select regions of the developing mouse nervous system. Typically Slits signal through the Robo receptors, however they also have Robo-independent functions. The novel Slit receptor Eva-1, recently discovered in C. elegans, and the human orthologue of which is located in the Down syndrome critical region on chromosome 21, could account for some of these Robo independent functions as well as provide selectivity to Robo-mediated axon responses to Slit. Here we investigate the expression of the mammalian orthologue EVA1C in regions of the developing mouse nervous system which have been shown to exhibit Robo-dependent and -independent responses to Slit. We report that EVA1C is expressed by axons contributing to commissures, tracts and nerve pathways of the developing spinal cord and forebrain. Furthermore it is expressed by axons that display both Robo-dependent and -independent functions of Slit, supporting a role for EVA1C in Slit/Robo mediated neural circuit formation in the developing nervous system.
Collapse
Affiliation(s)
- Gregory James
- School of Biomedical Science, University of Queensland, Brisbane, Australia
| | - Simon R. Foster
- School of Biomedical Science, University of Queensland, Brisbane, Australia
| | - Brian Key
- School of Biomedical Science, University of Queensland, Brisbane, Australia
- * E-mail: (BK); (AB)
| | - Annemiek Beverdam
- School of Biomedical Science, University of Queensland, Brisbane, Australia
- * E-mail: (BK); (AB)
| |
Collapse
|
24
|
Down M, Willshaw DA, Pratt T, Price DJ. Steerable-filter based quantification of axonal populations at the developing optic chiasm reveal significant defects in Slit2(-/-) as well as Slit1(-/-)Slit2(-/-) embryos. BMC Neurosci 2013; 14:9. [PMID: 23320558 PMCID: PMC3579723 DOI: 10.1186/1471-2202-14-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 01/12/2013] [Indexed: 11/15/2022] Open
Abstract
Background Previous studies have suggested that the axon guidance proteins Slit1 and Slit2 co-operate to establish the optic chiasm in its correct position at the ventral diencephalic midline. This is based on the observation that, although both Slit1 and Slit2 are expressed around the ventral midline, mice defective in either gene alone exhibit few or no axon guidance defects at the optic chiasm whereas embryos lacking both Slit1 and Slit2 develop a large additional chiasm anterior to the chiasm’s normal position. Here we used steerable-filters to quantify key properties of the population of axons at the chiasm in wild-type, Slit1−/−, Slit2−/− and Slit1−/−Slit2−/− embryos. Results We applied the steerable-filter algorithm successfully to images of embryonic retinal axons labelled from a single eye shortly after they have crossed the midline. We combined data from multiple embryos of the same genotype and made statistical comparisons of axonal distributions, orientations and curvatures between genotype groups. We compared data from the analysis of axons with data on the expression of Slit1 and Slit2. The results showed a misorientation and a corresponding anterior shift in the position of many axons at the chiasm of both Slit2−/− and Slit1−/−Slit2−/− mutants. There were very few axon defects at the chiasm of Slit1−/− mutants. Conclusions We found defects of the chiasms of Slit1−/−Slit2−/− and Slit1−/− mutants similar to those reported previously. In addition, we discovered previously unreported defects resulting from loss of Slit2 alone. This indicates the value of a quantitative approach to complex pathway analysis and shows that Slit2 can act alone to control aspects of retinal axon routing across the ventral diencephalic midline.
Collapse
Affiliation(s)
- Matthew Down
- Institute for Adaptive and Neural Computation, University of Edinburgh, Edinburgh, UK
| | | | | | | |
Collapse
|
25
|
Schwend T, Lwigale PY, Conrad GW. Nerve repulsion by the lens and cornea during cornea innervation is dependent on Robo-Slit signaling and diminishes with neuron age. Dev Biol 2012; 363:115-27. [PMID: 22236962 PMCID: PMC3288411 DOI: 10.1016/j.ydbio.2011.12.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 12/15/2011] [Accepted: 12/16/2011] [Indexed: 11/29/2022]
Abstract
The cornea, the most densely innervated tissue on the surface of the body, becomes innervated in a series of highly coordinated developmental events. During cornea development, chick trigeminal nerve growth cones reach the cornea margin at embryonic day (E)5, where they are initially repelled for days from E5 to E8, instead encircling the corneal periphery in a nerve ring prior to entering on E9. The molecular events coordinating growth cone guidance during cornea development are poorly understood. Here we evaluated a potential role for the Robo-Slit nerve guidance family. We found that Slits 1, 2 and 3 expression in the cornea and lens persisted during all stages of cornea innervation examined. Robo1 expression was developmentally regulated in trigeminal cell bodies, expressed robustly during nerve ring formation (E5-8), then later declining concurrent with projection of growth cones into the cornea. In this study we provide in vivo and in vitro evidence that Robo-Slit signaling guides trigeminal nerves during cornea innervation. Transient, localized inhibition of Robo-Slit signaling, by means of beads loaded with inhibitory Robo-Fc protein implanted into the developing eyefield in vivo, led to disorganized nerve ring formation and premature cornea innervation. Additionally, when trigeminal explants (source of neurons) were oriented adjacent to lens vesicles or corneas (source of repellant molecules) in organotypic tissue culture both lens and cornea tissues strongly repelled E7 trigeminal neurites, except in the presence of inhibitory Robo-Fc protein. In contrast, E10 trigeminal neurites were not as strongly repelled by cornea, and presence of Robo-Slit inhibitory protein had no effect. In full, these findings suggest that nerve repulsion from the lens and cornea during nerve ring formation is mediated by Robo-Slit signaling. Later, a shift in nerve guidance behavior occurs, in part due to molecular changes in trigeminal neurons, including Robo1 downregulation, thus allowing nerves to find the Slit-expressing cornea permissive for growth cones.
Collapse
Affiliation(s)
- Tyler Schwend
- Division of Biology, Ackert Hall, Kansas State University, Manhattan, KS 66506, USA.
| | | | | |
Collapse
|
26
|
Lee S, Kim JH, Kim JH, Seo JW, Han HS, Lee WH, Mori K, Nakao K, Barasch J, Suk K. Lipocalin-2 Is a chemokine inducer in the central nervous system: role of chemokine ligand 10 (CXCL10) in lipocalin-2-induced cell migration. J Biol Chem 2011; 286:43855-43870. [PMID: 22030398 DOI: 10.1074/jbc.m111.299248] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The secreted protein lipocalin-2 (LCN2) has been implicated in diverse cellular processes, including cell morphology and migration. Little is known, however, about the role of LCN2 in the CNS. Here, we show that LCN2 promotes cell migration through up-regulation of chemokines in brain. Studies using cultured glial cells, microvascular endothelial cells, and neuronal cells suggest that LCN2 may act as a chemokine inducer on the multiple cell types in the CNS. In particular, up-regulation of CXCL10 by JAK2/STAT3 and IKK/NF-κB pathways in astrocytes played a pivotal role in LCN2-induced cell migration. The cell migration-promoting activity of LCN2 in the CNS was verified in vivo using mouse models. The expression of LCN2 was notably increased in brain following LPS injection or focal injury. Mice lacking LCN2 showed the impaired migration of astrocytes to injury sites with a reduced CXCL10 expression in the neuroinflammation or injury models. Thus, the LCN2 proteins, secreted under inflammatory conditions, may amplify neuroinflammation by inducing CNS cells to secrete chemokines such as CXCL10, which recruit additional inflammatory cells.
Collapse
Affiliation(s)
- Shinrye Lee
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jong-Heon Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jae-Hong Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jung-Wan Seo
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Hyung-Soo Han
- Department of Physiology, Brain Science & Engineering Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Won-Ha Lee
- Departments of School of Life Sciences and Biotechnology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Kiyoshi Mori
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kazuwa Nakao
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Jonathan Barasch
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10027
| | - Kyoungho Suk
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea.
| |
Collapse
|
27
|
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
|
28
|
Alajez NM, Lenarduzzi M, Ito E, Hui ABY, Shi W, Bruce J, Yue S, Huang SH, Xu W, Waldron J, O'Sullivan B, Liu FF. MiR-218 suppresses nasopharyngeal cancer progression through downregulation of survivin and the SLIT2-ROBO1 pathway. Cancer Res 2011; 71:2381-91. [PMID: 21385904 DOI: 10.1158/0008-5472.can-10-2754] [Citation(s) in RCA: 229] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Nasopharayngeal carcinoma (NPC) is an Epstein-Barr virus-associated malignancy most common in East Asia and Africa. Here we report frequent downregulation of the microRNA miR-218 in primary NPC tissues and cell lines where it plays a critical role in NPC progression. Suppression of miR-218 was associated with epigenetic silencing of SLIT2 and SLIT3, ligands of ROBO receptors that have been previously implicated in tumor angiogenesis. Exogenous expression of miR-218 caused significant toxicity in NPC cells in vitro and delayed tumor growth in vivo. We used an integrated trimodality approach to identify targets of miR-218 in NPC, cervical, and breast cell lines. Direct interaction between miR-218 and the 3'-untranslated regions (UTR) of mRNAs encoding ROBO1, survivin (BIRC5), and connexin43 (GJA1) was validated in a luciferase-based transcription reporter assay. Mechanistic investigations revealed a negative feedback loop wherein miR-218 regulates NPC cell migration via the SLIT-ROBO pathway. Pleotropic effects of miR-218 on NPC survival and migration were rescued by enforced expression of miR-218-resistant, engineered isoforms of survivin and ROBO1, respectively. In clinical specimens of NPC (n=71), ROBO1 overexpression was significantly associated with worse overall (P=0.04, HR=2.4) and nodal relapse-free survival (P=0.008, HR=6.0). Our findings define an integrative tumor suppressor function for miR-218 in NPC and further suggest that restoring miR-218 expression in NPC might be useful for its clinical management.
Collapse
Affiliation(s)
- Nehad M Alajez
- Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
The failure of the optic nerve to regenerate after injury or in neurodegenerative disease remains a major clinical and scientific problem. Retinal ganglion cell (RGC) axons course through the optic nerve and carry all the visual information to the brain, but after injury, they fail to regrow through the optic nerve and RGC cell bodies typically die, leading to permanent loss of vision. There are at least 4 hurdles to overcome in preserving RGCs and regenerating their axons: 1) increase RGC survival, 2) overcome the inhibitory environment of the optic nerve, 3) enhance RGC intrinsic axon growth potential, and 4) optimize the mapping of RGC connections back into their targets in the brain.
Collapse
|
30
|
Torre ER, Gutekunst CA, Gross RE. Expression by midbrain dopamine neurons of Sema3A and 3F receptors is associated with chemorepulsion in vitro but a mild in vivo phenotype. Mol Cell Neurosci 2010; 44:135-53. [PMID: 20298787 PMCID: PMC2862895 DOI: 10.1016/j.mcn.2010.03.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 02/17/2010] [Accepted: 03/03/2010] [Indexed: 12/23/2022] Open
Abstract
Here we explore the role of semaphorin 3A and 3F (Sema3A, Sema3F) in the formation of the mesotelencephalic pathway. We show that Sema3A and 3F are expressed in the ventral mesencephalon (VM) of E13.5 rat embryos; the receptors Neuropilin 1 and Neuropilin 2, and co-receptors L1CAM, NrCAM, and Plexins A1 and A3 but not A4 are expressed by VM dopaminergic neurons; these neurons bind Sema3A and 3F in vitro which induces collapse of their growth cones and elicits, with different potencies, a repulsive response; and this response is absent in axons from Nrp1 and Nrp2 null embryos. Despite these in vitro effects, only very mild anatomical defects were detected in the organization of the mesotelencephalic pathway in embryonic and adult Nrp1 or Nrp2 null mice. However, the dopaminergic meso-habenular pathway and catecholaminergic neurons in the parafascicular and paraventricular nuclei of the thalamus were significantly affected in Nrp2 null mice. These data are consistent with a model whereby Sema3A and 3F, in combination with other guidance molecules, contributes to the navigation of DA axons to their final synaptic targets.
Collapse
Affiliation(s)
- Enrique R. Torre
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
| | | | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
- Department of Neurology and Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
31
|
Hocking JC, Hehr CL, Bertolesi GE, Wu JY, McFarlane S. Distinct roles for Robo2 in the regulation of axon and dendrite growth by retinal ganglion cells. Mech Dev 2009; 127:36-48. [PMID: 19961927 DOI: 10.1016/j.mod.2009.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 11/18/2009] [Accepted: 11/27/2009] [Indexed: 11/28/2022]
Abstract
Guidance factors act on the tip of a growing axon to direct it to its target. What role these molecules play, however, in the control of the dendrites that extend from that axon's cell body is poorly understood. Slits, through their Robo receptors, guide many types of axons, including those of retinal ganglion cells (RGCs). Here we assess and contrast the role of Slit/Robo signalling in the growth and guidance of the axon and dendrites extended by RGCs in Xenopus laevis. As Xenopus RGCs extend dendrites, they express robo2 and robo3, while slit1 and slit2 are expressed in RGCs and in the adjacent inner nuclear layer. Interestingly, our functional data with antisense knockdown and dominant negative forms of Robo2 (dnRobo2) and Robo3 (dnRobo3) indicate that Slit/Robo signalling has no role in RGC dendrite guidance, and instead is necessary to stimulate dendrite branching, primarily via Robo2. Our in vitro culture data argue that Slits are the ligands involved. In contrast, both dnRobo2 and dnRobo3 inhibited the extension of axons and caused the misrouting of some axons. Based on these data, we propose that Robo signalling can have distinct functions in the axon and dendrites of the same cell, and that the specific combinations of Robo receptors could underlie these differences. Slit acts via Robo2 in dendrites as a branching/growth factor but not in guidance, while Robo2 and Robo3 function in concert in axons to mediate axonal interactions and respond to Slits as guidance factors. These data underscore the likelihood that a limited number of extrinsic factors regulate the distinct morphologies of axons and dendrites.
Collapse
|
32
|
Huang L, Xu Y, Yu W, Li X, Liqun C, He X, Peiying H. Robo1: a potential role in ocular angiogenesis. Curr Eye Res 2009; 34:1019-29. [PMID: 19958120 DOI: 10.3109/02713680903308495] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Robo1, a member of the roundabout (Robo) family, serves as neuronal guidance receptors and has been reported to mediate tumor angiogenesis. In this study, we investigated the function of Robo1 and its possible role in retinal angiogenesis in vitro and in vivo. METHODS Expression of Robo1 in relation to retinal neovascularization was studied in an animal model of retinopathy of prematurity (ROP). siRNA technology was used to knockdown Robo1 expression to study its effects on monkey choroidal retinal endothelial cells (RF/6A) in vitro. Cell proliferation, migration, spreading, cycling, and apoptosis were assessed with the methyl thiazolyl tetrazolium (MTT), Boyden chamber, immunohistochemistry, and flow cytometry assay. Tube formation by RF/6A on Matrigel was also analyzed. RESULTS Robo1 expression was elevated in the ROP murine eyes (P < 0.01). Knockdown of Robo1 expression inhibited cell proliferation and migration. Tube formation by RF/6A was also disturbed. CONCLUSIONS Our results show that Robo1 inhibits choroidal and retinal angiogenesis in vitro. Further studies are ongoing to evaluate Robo1 as a potential target for the treatment of choroidal or retinal angiogenesis.
Collapse
Affiliation(s)
- Lvzhen Huang
- Department of Ophthalmology, Peking University People's Hospital, Xizhimen South Street 11, Xi Cheng District, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
33
|
Robo2 is required for Slit-mediated intraretinal axon guidance. Dev Biol 2009; 335:418-26. [PMID: 19782674 PMCID: PMC2814049 DOI: 10.1016/j.ydbio.2009.09.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 09/21/2009] [Accepted: 09/21/2009] [Indexed: 11/21/2022]
Abstract
The developing optic pathway has proven one of the most informative model systems for studying mechanisms of axon guidance. The first step in this process is the directed extension of retinal ganglion cell (RGC) axons within the optic fibre layer (OFL) of the retina towards their exit point from the eye, the optic disc. Previously, we have shown that the inhibitory guidance molecules, Slit1 and Slit2, regulate two distinct aspects of intraretinal axon guidance in a region-specific manner. Using knockout mice, we have found that both of these guidance activities are mediated via Robo2. Of the four vertebrate Robos, only Robo1 and Robo2 are expressed by RGCs. In mice lacking robo1 intraretinal axon guidance occurs normally. However, in mice lacking robo2 RGC axons make qualitatively and quantitatively identical intraretinal pathfinding errors to those reported previously in Slit mutants. This demonstrates clearly that, as in other regions of the optic pathway, Robo2 is the major receptor required for intraretinal axon guidance. Furthermore, the results suggest strongly that redundancy with other guidance signals rather than different receptor utilisation is the most likely explanation for the regional specificity of Slit function during intraretinal axon pathfinding.
Collapse
|
34
|
Braisted JE, Ringstedt T, O'Leary DDM. Slits are chemorepellents endogenous to hypothalamus and steer thalamocortical axons into ventral telencephalon. Cereb Cortex 2009; 19 Suppl 1:i144-51. [PMID: 19435711 PMCID: PMC2693534 DOI: 10.1093/cercor/bhp035] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Thalamocortical axons (TCAs) originate in dorsal thalamus, extend ventrally along the lateral thalamic surface, and as they approach hypothalamus make a lateral turn into ventral telencephalon. In vitro studies show that hypothalamus releases a chemorepellent for TCAs, and analyses of knockout mice indicate that Slit chemorepellents and their receptor Robo2 influence TCA pathfinding. We show that Slit chemorepellents are the hypothalamic chemorepellent and act through Robos to steer TCAs into ventral telencephalon. During TCA pathfinding, Slit1 and Slit2 are expressed in hypothalamus and ventral thalamus and Robo1 and Robo2 are expressed in dorsal thalamus. In collagen gel cocultures of dorsal thalamus and Slit2-expressing cells, axon number and length are decreased on the explant side facing Slit2-expressing cells, overall axon outgrowth is diminished, and axons turn away from the Slit2-expressing cells. Thus, Slit2 is an inhibitor and chemorepellent for dorsal thalamic axons. Collagen gel cocultures of dorsal thalamus with sections of live diencephalon, with and without the hypothalamus portion overlaid with Robo2-fc-expressing cells to block Slit function, identify Slits as the hypothalamic chemorepellent. Thus, Slits are chemorepellents for TCAs endogenous to hypothalamus and steer TCAs from diencephalon into ventral telencephalon, a critical pathfinding event defective in Slit and Robo2 mutant mice.
Collapse
Affiliation(s)
- Janet E Braisted
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
35
|
Huang L, Yu W, Li X, Niu L, Li K, Li J. Robo1/robo4: different expression patterns in retinal development. Exp Eye Res 2009; 88:583-8. [PMID: 19084519 DOI: 10.1016/j.exer.2008.11.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 10/29/2008] [Accepted: 11/13/2008] [Indexed: 01/30/2023]
Abstract
Two members of the roundabout (Robo) family, Robo1 and Robo4, serve as neuronal guidance receptors. During neurogenesis, Robo1 and Robo4 participate in axonal guidance by mediating a repulsive signal. It has been reported that Robo4 is mainly expressed in the vasculature and that Robo1 is expressed both in neural and non-neural tissues. However, the roles of these Robo proteins in the mammalian vasculature are still unclear. In this current study, the expression patterns of Robo1 and Robo4 in the retinal vasculature were determined using C57BL/6J mice at postnatal days (P) 1, P3, P5, P7, P9, P12, P14, P17, P21 and adult mice (1month). We found that Robo4 was expressed not only in the retinal vessels but also in the retinal ganglion cell and photoreceptor layers during retinal development. Robo4 expression peaked at P3 and P9, which suggest that Robo4 may function in stabilizing the retinal vasculature. Robo1 expression was observed in the retina neuronal cells and vessels. Both Robo1 mRNA and protein expression showed a typical expression pattern, which related to Robo1's roles in the different stages of retinal vascular development in the murine retina. Robo1 displayed high expression levels at P1 (correlated with superficial vascular plexus formation) and P7 (correlated with deep vascular plexus formation). The high levels of Robo1 during these two well-defined phases of retinal capillary plexus formation indicate that Robo1 is likely to play a part in retinal neovascularization.
Collapse
Affiliation(s)
- Lvzhen Huang
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Slit was identified in Drosophila embryo as a gene involved in the patterning of larval cuticle. It was later shown that Slit is synthesized in the fly central nervous system by midline glia cells. Slit homologues have since been found in C. elegans and many vertebrate species, from amphibians, fishes, birds to mammals. A single slit was isolated in invertebrates, whereas there are three slit genes (slit1-slit3) in mammals, that have around 60% homology. All encodes large ECM glycoproteins of about 200 kDa (Fig. 1A), comprising, from their N terminus to their C terminus, a long stretch of four leucine rich repeats (LRR) connected by disulphide bonds, seven to nine EGF repeats, a domain, named ALPS (Agrin, Perlecan, Laminin, Slit) or laminin G-like module (see ref 17), and a cystein knot (Fig. 1A). Alternative spliced transcripts have been reported for Drosophila Slit2, human Slit2 and Slit3, and Slit1. Moreover, two Slit1 isoforms exist in zebrafish as a consequence of gene duplication. Last, in mammals, two Slit2 isoforms can be purified from brain extracts, a long 200 kDa one and a shorter 150 kDa form (Slit2-N) that was shown to result from the proteolytic processing of full-length Slit2. Human Slit and Slit3 and Drosophila Slit are also cleaved by an unknown protease in a large N-terminal fragment and a shorter C-terminal fragment, suggesting conserved mechanisms for Slit cleavage across species. Moreover, Slit fragments have different cell association characteristics in cell culture suggesting that they may also have different extents of diffusion, different binding properties, and, hence, different functional activities in vivo. This conclusion is supported by in vitro data showing that full-length Slit2 functions as an antagonist of Slit2-N in the DRG branching assay, and that Slit2-N, not full-length Slit2, causes collapse of OB growth cones. In addition, Slit1-N and full-length Slit1 can induce branching of cortical neurons (see below), but only full-length Slit1 repels cortical axons. Structure-function analysis in vertebrates and Drosophila demonstrated that the LRRs of Slits are required and sufficient to mediate their repulsive activities in neurons. More recent detailed structure function analysis of the LRR domains of Drosophila Slit, revealed that the active site of Slit (at least regarding its pro-angiogenic activity) is located on the second of the fourth LRR (LRR2), which is highly conserved between Slits. Slit can also dimerize through the LRR4 domain and the cystein knot.However, a Slit1 spliced-variant that lacks the cysteine knot and does not dimerize is still able to repel OB axons.
Collapse
|
37
|
Tian NM, Pratt T, Price DJ. Foxg1 regulates retinal axon pathfinding by repressing an ipsilateral program in nasal retina and by causing optic chiasm cells to exert a net axonal growth-promoting activity. Development 2008; 135:4081-9. [PMID: 19004857 PMCID: PMC6207343 DOI: 10.1242/dev.023572] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mammalian binocular vision relies on the divergence of retinal ganglion cell axons at the optic chiasm, with strictly controlled numbers projecting contralaterally and ipsilaterally. In mouse, contralateral projections arise from the entire retina, whereas ipsilateral projections arise from ventrotemporal retina. We investigate how development of these patterns of projection is regulated by the contralateral determinant Foxg1, a forkhead box transcription factor expressed in nasal retina and at the chiasm. In nasal retina, loss of Foxg1 causes increased numbers of ipsilateral projections and ectopic expression of the ipsilateral determinants Zic2, Ephb1 and Foxd1, indicating that nasal retina is competent to express an ipsilateral program that is normally suppressed by Foxg1. Using co-cultures that combine Foxg1-expressing with Foxg1-null retinal explants and chiasm cells, we provide functional evidence that Foxg1 promotes contralateral projections through actions in nasal retina, and that in chiasm cells, Foxg1 is required for the generation of a hitherto unrecognized activity supporting RGC axon growth.
Collapse
Affiliation(s)
- Natasha M. Tian
- Centre for Integrative Physiology, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Thomas Pratt
- Centre for Integrative Physiology, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - David J. Price
- Centre for Integrative Physiology, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
38
|
Sánchez-Camacho C, Bovolenta P. Autonomous and non-autonomous Shh signalling mediate the in vivo growth and guidance of mouse retinal ganglion cell axons. Development 2008; 135:3531-41. [PMID: 18832395 DOI: 10.1242/dev.023663] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In non-mammalian vertebrates, the relatively homogeneous population of retinal ganglion cells (RGCs) differentiates and projects entirely to the contralateral side of the brain under the influence of sonic hedgehog (Shh). In mammals, by contrast, there are two different RGC types: the Zic2-positive ipsilateral projecting and the Isl2-positive contralateral projecting. We asked whether the axons of these two populations respond to Shh and if their response differs. We have also analysed whether midline- and RGC-derived Shh contributes to the growth of the axons in the proximal visual pathway. We show that these two RGC types are characterised by a differential expression of Shh signalling components and that they respond differently to Shh when challenged in vitro. In vivo blockade of Shh activity, however, alters the path and distribution mostly of the contralateral projecting RGC axons at the chiasm, indicating that midline-derived Shh participates in funnelling contralateral visual fibres in this region. Furthermore, interference with Shh signalling in the RGCs themselves causes abnormal growth and navigation of contralateral projecting axons in the proximal portion of the pathway, highlighting a novel cell-autonomous mechanism by which Shh can influence growth cone behaviour.
Collapse
Affiliation(s)
- Cristina Sánchez-Camacho
- Departamento de Neurobiología Molecular Celular y del Desarrollo, Instituto Cajal, CSIC and CIBER de Enfermedades Raras (CIBERER) 37, Madrid 28002, Spain
| | | |
Collapse
|
39
|
Plachez C, Andrews W, Liapi A, Knoell B, Drescher U, Mankoo B, Zhe L, Mambetisaeva E, Annan A, Bannister L, Parnavelas JG, Richards LJ, Sundaresan V. Robos are required for the correct targeting of retinal ganglion cell axons in the visual pathway of the brain. Mol Cell Neurosci 2008; 37:719-30. [PMID: 18272390 DOI: 10.1016/j.mcn.2007.12.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 11/24/2007] [Accepted: 12/12/2007] [Indexed: 11/26/2022] Open
Abstract
Axonal projections from the retina to the brain are regulated by molecules including the Slit family of ligands [Thompson, H., Barker, D., Camand, O., Erskine, L., 2006a. Slits contribute to the guidance of retinal ganglion cell axons in the mammalian optic tract. Dev. Biol. 296, 476-484, Thompson, H., Camand, O., Barker, D., Erskine, L., 2006b. Slit proteins regulate distinct aspects of retinal ganglion cell axon guidance within dorsal and ventral retina. J. Neurosci. 26, 8082-8091]. However, the roles of Slit receptors in mammals, (termed Robos), have not been investigated in visual system development. Here we examined Robo1 and 2 mutant mice and found that Robos regulate the correct targeting of retinal ganglion cell (RGC) axons along the entire visual projection. We noted aberrant projections of RGC axons into the cerebral cortex, an area not normally targeted by RGC axons. The optic chiasm was expanded along the rostro-caudal axis (similar to Slit mutant mice, Plump, A.S., Erskine, L., Sabatier, C., Brose, K., Epstein, C.J., Goodman, C.S., Mason, C.A., Tessier-Lavigne, M., 2002. Slit1 and Slit2 cooperate to prevent premature midline crossing of retinal axons in the mouse visual system. Neuron 33, 219-232), with ectopic crossing points, and some axons projecting caudally toward the corticospinal tract. Further, we found that axons exuberantly projected into the diencephalon. These defects were more pronounced in Robo2 than Robo1 knockout animals, implicating Robo2 as the predominant Robo receptor in visual system development.
Collapse
Affiliation(s)
- Céline Plachez
- The University of Maryland, Baltimore, School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bao ZZ. Intraretinal projection of retinal ganglion cell axons as a model system for studying axon navigation. Brain Res 2008; 1192:165-77. [PMID: 17320832 PMCID: PMC2267003 DOI: 10.1016/j.brainres.2007.01.116] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2006] [Revised: 01/24/2007] [Accepted: 01/26/2007] [Indexed: 01/19/2023]
Abstract
The initial step of retinal ganglion cell (RGC) axon pathfinding involves directed growth of RGC axons toward the center of the retina, the optic disc, a process termed "intraretinal guidance". Due to the accessibility of the system, and with various embryological, molecular and genetic approaches, significant progress has been made in recent years toward understanding the mechanisms involved in the precise guidance of the RGC axons. As axons are extending from RGCs located throughout the retina, a multitude of factors expressed along with the differentiation wave are important for the guidance of the RGC axons. To ensure that the RGC axons are oriented correctly, restricted to the optic fiber layer (OFL) of the retina, and exit the eye properly, different sets of positive and negative factors cooperate in the process. Fasciculation mediated by a number of cell adhesion molecules (CAMs) and modulation of axonal response to guidance factors provide additional mechanisms to ensure proper guidance of the RGC axons. The intraretinal axon guidance thus serves as an excellent model system for studying how different signals are regulated, modulated and integrated for guiding a large number of axons in three-dimensional space.
Collapse
Affiliation(s)
- Zheng-Zheng Bao
- Department of Medicine and Cell Biology, Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
41
|
Curinga G, Smith GM. Molecular/genetic manipulation of extrinsic axon guidance factors for CNS repair and regeneration. Exp Neurol 2008; 209:333-42. [PMID: 17706643 PMCID: PMC2255571 DOI: 10.1016/j.expneurol.2007.06.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 06/25/2007] [Accepted: 06/25/2007] [Indexed: 11/22/2022]
Abstract
During development, guidance molecules play a key role in the formation of complex circuits required for neural functions. With the cessation of development, this exuberant growth process slows and stabilizes, and inhibitory molecules expressed by glia prevent initial attempts for axonal regeneration. In this review, we discuss the expression patterns and relative contribution of several guidance molecules on the regenerative process. Injury to the immature CNS or species capable of regenerating exhibit a complete or partial recapitulation of their developmental guidance patterns, whereas similar injuries to adult mammals results in altered expression that acts to further hinder regeneration. Manipulations of guidance molecules after injury have been used to control detrimental effects of axon sprouting and target regenerating axons within the spinal cord.
Collapse
Affiliation(s)
- Gabrielle Curinga
- Department of Physiology and Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA
| | | |
Collapse
|
42
|
Yao Q, Jin WL, Wang Y, Ju G. Regulated shuttling of Slit-Robo-GTPase activating proteins between nucleus and cytoplasm during brain development. Cell Mol Neurobiol 2008; 28:205-21. [PMID: 17710530 PMCID: PMC11514978 DOI: 10.1007/s10571-007-9187-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Accepted: 07/28/2007] [Indexed: 12/11/2022]
Abstract
(1) A Little information exists on the distribution of Slit-Robo-GTPase activating proteins (srGAPs), particularly about their intracellular locations, which may provide further clues to their functions. The purpose of this study is to elucidate the expression patterns of the three srGAPs in wild-type rat brains at adult and various developmental stages, and in the cultured cortical neurons. (2) Immunohistochemical method was applied to detect the distribution and localization of the srGAPs in the normal rat brains at adult and various developmental stages, and in the cultured cortical neurons using the rabbit polyclonal antibodies. (3) Immunohistochemical analysis demonstrated that the three srGAPs were mainly expressed in neurons throughout the brain. More importantly, srGAPs translocated during development by a highly regulated shuttling process between the nucleus and the cytoplasm of neurons and their expression patterns were not overlapping. In cultured cortical neurons srGAPs were found in equal amounts in the cytoplasm, nucleus, in neurites, and growth cones. When neurons were maintained in vitro for longer time, the amount of srGAPs in the nucleus strongly increased. (4) These results suggest that srGAPs are not only involved in the regulation of the Slit-Robo signal transduction, but also in neuronal development and that the translocation of srGAPs is important for their functions.
Collapse
Affiliation(s)
- Qin Yao
- Institute of Neurosciences, the Fourth Military Medical University, 17 Chang Le Xi Road, Xi’an, 710032 P.R. China
| | - Wei-Lin Jin
- Institute of Neurosciences, Shanghai JiaoTong University, 800 Dongchuan Road, MinHang, Shanghai 200240 P.R. China
| | - Ying Wang
- Institute of Neurosciences, the Fourth Military Medical University, 17 Chang Le Xi Road, Xi’an, 710032 P.R. China
| | - Gong Ju
- Institute of Neurosciences, the Fourth Military Medical University, 17 Chang Le Xi Road, Xi’an, 710032 P.R. China
- Institute of Neurosciences, Shanghai JiaoTong University, 800 Dongchuan Road, MinHang, Shanghai 200240 P.R. China
| |
Collapse
|
43
|
Rüdiger T, Bolz J. Acetylcholine influences growth cone motility and morphology of developing thalamic axons. Cell Adh Migr 2008; 2:30-7. [PMID: 19262162 DOI: 10.4161/cam.2.1.5909] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The neurotransmitter acetylcholine (ACh) is expressed in the developing telencephalon at the time when thalamic axons project to the cortex, long before synapses are being formed. Since previous studies demonstrated an influence of ACh on neurite extension we used different in vitro assays to examine possible effects of ACh on the growth of thalamic axons. In explant cultures, application of ACh reduced the length of thalamic axons in a dose dependent manner, an effect that could also be evoked by selective muscarinic and nicotinic agonists. Time-lapse imaging of thalamic axons exposed to microscopic gradients of ACh revealed that growth cones no longer advanced, but maintained high filopodial activity. This growth cone pausing was not accompanied by axon retraction or growth cone collapse. It could at least partially be blocked by muscarinic and nicotinic antagonists, indicating that both types of ACh receptors contribute to mediate these effects on thalamic axons. Finally, we also found that ACh changed the morphology of growth cones; they became larger and extended more filopodia. Since such changes in the structure and motility of growth cones are observed at decision regions along the path of many fiber populations including thalamic axons, we suggest that ACh plays a role during the elaboration of thalamocortical projections.
Collapse
Affiliation(s)
- Tina Rüdiger
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, Jena, Germany
| | | |
Collapse
|
44
|
Kapfhammer JP, Xu H, Raper JA. The detection and quantification of growth cone collapsing activities. Nat Protoc 2007; 2:2005-11. [PMID: 17703212 DOI: 10.1038/nprot.2007.295] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Growth cone guidance during development, as well as axonal extension in neural repair and plasticity, is strongly regulated by both attractive (growth-promoting) and repulsive (growth-inhibiting) guidance molecules. The growth cone collapse assay has been widely and successfully used for the identification and purification of molecules that are repulsive to growth cones or inhibit axonal outgrowth. Here we provide a detailed description of the assay, which uses the morphology of the growth cone after exposure to a test protein as the readout. With the modifications detailed in this protocol, this assay can be used for the biochemical enrichment of proteins with a collapsing activity and for the identification of a collapsing activity of a known protein or gene. This assay does not require very specialized equipment and can be established by every lab with experience in neuronal cell culture. It can be completed in 3 d.
Collapse
Affiliation(s)
- Josef P Kapfhammer
- Anatomisches Institut, Universität Basel, Pestalozzistr. 20, CH-4056 Basel, Switzerland.
| | | | | |
Collapse
|
45
|
Keizer-Gunnink I, Kortholt A, Van Haastert PJM. Chemoattractants and chemorepellents act by inducing opposite polarity in phospholipase C and PI3-kinase signaling. ACTA ACUST UNITED AC 2007; 177:579-85. [PMID: 17517960 PMCID: PMC2064204 DOI: 10.1083/jcb.200611046] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During embryonic development, cell movement is orchestrated by a multitude of attractants and repellents. Chemoattractants applied as a gradient, such as cAMP with Dictyostelium discoideum or fMLP with neutrophils, induce the activation of phospholipase C (PLC) and phosphoinositide 3 (PI3)-kinase at the front of the cell, leading to the localized depletion of phosphatidylinositol 4,5-bisphosphate (PI[4,5]P2) and the accumulation of phosphatidylinositol-3,4,5-trisphosphate (PI[3,4,5]P3). Using D. discoideum, we show that chemorepellent cAMP analogues induce localized inhibition of PLC, thereby reversing the polarity of PI(4,5)P2. This leads to the accumulation of PI(3,4,5)P3 at the rear of the cell, and chemotaxis occurs away from the source. We conclude that a PLC polarity switch controls the response to attractants and repellents.
Collapse
Affiliation(s)
- Ineke Keizer-Gunnink
- Department of Molecular Cell Biology, University of Groningen, Haren, Netherlands
| | | | | |
Collapse
|
46
|
Erskine L, Herrera E. The retinal ganglion cell axon's journey: insights into molecular mechanisms of axon guidance. Dev Biol 2007; 308:1-14. [PMID: 17560562 DOI: 10.1016/j.ydbio.2007.05.013] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 05/02/2007] [Accepted: 05/10/2007] [Indexed: 02/02/2023]
Abstract
The developing visual system has proven to be one of the most informative models for studying axon guidance decisions. The pathway is composed of the axons of a single neuronal cell type, the retinal ganglion cell (RGC), that navigate through a series of intermediate targets on route to their final destination. The molecular basis of optic pathway development is beginning to be elucidated with cues such as netrins, Slits and ephrins playing a key role. Other factors best characterised for their role as morphogens in patterning developing tissues, such as sonic hedgehog (Shh) and Wnts, also act directly on RGC axons to influence guidance decisions. The transcriptional basis of the spatial-temporal expression of guidance cues and their cognate receptors within the developing optic pathway as well as mechanisms underlying the plasticity of guidance responses also are starting to be understood. This review will focus on our current understanding of the molecular mechanisms directing the early development of functional connections in the developing visual system and the insights these studies have provided into general mechanisms of axon guidance.
Collapse
Affiliation(s)
- Lynda Erskine
- Division of Visual Science, Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | | |
Collapse
|
47
|
de Wit J, Verhaagen J. Proteoglycans as modulators of axon guidance cue function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 600:73-89. [PMID: 17607948 DOI: 10.1007/978-0-387-70956-7_7] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Organizing a functional neuronal network requires the precise wiring of neuronal connections. In order to find their correct targets, growth cones navigate through the extracellular matrix guided by secreted and membrane-bound molecules of the slit, netrin, ephrin and semaphorin families. Although many of these axon guidance molecules are able to bind to heparan sulfate proteoglycans, the role of proteoglycans in regulating axon guidance cue function is only now beginning to be understood. Recent developmental studies in a wide range of model organisms have revealed a crucial role for heparan sulfate proteoglycans as modulators of key signaling pathways in axon guidance. In addition, emerging evidence indicates an essential role for chondroitin sulfate proteoglycans in modifying the guidance function of semaphorins. It is becoming increasingly clear that extracellular matrix molecules, rather than just constituting a structural scaffold, can critically influence axon guidance cue function in development, and may continue to do so in the injured adult nervous system.
Collapse
Affiliation(s)
- Joris de Wit
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, De Boelelaan 1087, 1081 HV Amsterdam, The Netherlands.
| | | |
Collapse
|
48
|
Conway C, Pratt T, Price D, Mason J. [P238]: Loss of heparan sulphate‐6‐sulphotransferase 1 results in retinal ganglion cell axon guidance errors at the optic chiasm and dorsal thalamus of the developing mouse visual system. Int J Dev Neurosci 2006. [DOI: 10.1016/j.ijdevneu.2006.09.297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
49
|
Lin L, Isacson O. Axonal growth regulation of fetal and embryonic stem cell-derived dopaminergic neurons by Netrin-1 and Slits. Stem Cells 2006; 24:2504-13. [PMID: 16840550 PMCID: PMC2613222 DOI: 10.1634/stemcells.2006-0119] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The physical restoration of dopamine circuits damaged or lost in Parkinson disease by implanting embryonic stem (ES)-derived cells may become a treatment. It is critical to understand responses of ES-derived dopamine (DA) neurons to guidance signals that determine axonal path and targeting. Using a collagen gel culture system, we examined effects of secreted molecules Netrin-1 and Slits on neurite outgrowth of fetal DA neurons and murine ES-differentiated DA neurons. We have previously shown that fetal DA neurons express DCC and Robo1/2 receptors and that Netrin-1 and Slit2 function as an attractant and a repellent for DA neurite outgrowth. In the present study, we observe that both Slit1 and Slit3 repel and inhibit neurite growth of fetal DA neurons. Here, we also demonstrate that ES-differentiated neurons including DA neurons express the Netrin receptor DCC and Slit receptor Robo proteins. In the gel culture system of ES cells, Netrin-1 promoted neurite outgrowth mediated by DCC receptor, and Slit1 and Slit3 were inhibitory for neurite outgrowth through Robo receptors. Slit2 appeared to exert inhibitory as well as repulsive effects in the coculture assay. However, unlike fetal DA neurites, no directed neurite outgrowth was observed in the cocultures of ES-derived DA neurons with Netrin-1-, Slit1-, and Slit3-producing cells. The findings suggest that ES-derived DA neurons generated by current protocols can respond to guidance cues in vitro in a similar manner to fetal cells but also exhibit distinct responses. This may result from developmental differences generated by present in vitro methods of cell patterning or conditioning during ES cell differentiation.
Collapse
Affiliation(s)
- Ling Lin
- Udall Parkinson's Disease Research Center of Excellence and Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, Massachusetts 02478, USA.
| | | |
Collapse
|
50
|
Niclou SP, Ehlert EME, Verhaagen J. Chemorepellent axon guidance molecules in spinal cord injury. J Neurotrauma 2006; 23:409-21. [PMID: 16629626 DOI: 10.1089/neu.2006.23.409] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Regenerating axons stop growing when they reach the border of the glial-fibrotic scar, presumably because they encounter a potent molecular barrier inhibiting growth cone advance. Chemorepulsive axon guidance molecules provide a non-permissive environment restricting and channeling axon growth in the developing nervous system. These molecules could also act as growth-inhibitory molecules in the regenerating nervous system. The receptors for repulsive guidance cues are expressed in the mature nervous system, suggesting that adult neurons are sensitive to the activity of developmentally active repulsive proteins. In this review, we summarize recent observations on semaphorins, ephrins, and slits in the injured brain and spinal cord, providing evidence that these proteins are major players in inhibiting axonal regeneration and establishing the glial-fibrotic scar.
Collapse
Affiliation(s)
- Simone P Niclou
- Netherlands Institute for Brain Research, Laboratory for Neuroregeneration, Amsterdam, The Netherlands.
| | | | | |
Collapse
|