1
|
Bu A, Afghah F, Castro N, Bawa M, Kohli S, Shah K, Rios B, Butty V, Raman R. Actuating Extracellular Matrices Decouple the Mechanical and Biochemical Effects of Muscle Contraction on Motor Neurons. Adv Healthc Mater 2025; 14:e2403712. [PMID: 39523700 PMCID: PMC11874633 DOI: 10.1002/adhm.202403712] [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: 09/28/2024] [Revised: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Emerging in vivo evidence suggests that repeated muscle contraction, or exercise, impacts peripheral nerves. However, the difficulty of isolating the muscle-specific impact on motor neurons in vivo, as well as the inability to decouple the biochemical and mechanical impacts of muscle contraction in this setting, motivates investigating this phenomenon in vitro. This study demonstrates that tuning the mechanical properties of fibrin enables longitudinal culture of highly contractile skeletal muscle monolayers, enabling functional characterization of and long-term secretome harvesting from exercised tissues. Motor neurons stimulated with exercised muscle-secreted factors significantly upregulate neurite outgrowth and migration, with an effect size dependent on muscle contraction intensity. Actuating magnetic microparticles embedded within fibrin hydrogels enable dynamically stretching motor neurons and non-invasively mimicking the mechanical effects of muscle contraction. Interestingly, axonogenesis is similarly upregulated in both mechanically and biochemically stimulated motor neurons, but RNA sequencing reveals different transcriptomic signatures between groups, with biochemical stimulation having a greater impact on cell signaling related to axonogenesis and synapse maturation. This study leverages actuating extracellular matrices to robustly validate a previously hypothesized role for muscle contraction in regulating motor neuron growth and maturation from the bottom-up through both mechanical and biochemical signaling.
Collapse
Affiliation(s)
- Angel Bu
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ferdows Afghah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Nicolas Castro
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Maheera Bawa
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sonika Kohli
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Karina Shah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Brandon Rios
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Vincent Butty
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ritu Raman
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
2
|
Desbois M, Grill B. Molecular regulation of axon termination in mechanosensory neurons. Development 2024; 151:dev202945. [PMID: 39268828 PMCID: PMC11698068 DOI: 10.1242/dev.202945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Spatially and temporally accurate termination of axon outgrowth, a process called axon termination, is required for efficient, precise nervous system construction and wiring. The mechanosensory neurons that sense low-threshold mechanical stimulation or gentle touch have proven exceptionally valuable for studying axon termination over the past 40 years. In this Review, we discuss progress made in deciphering the molecular and genetic mechanisms that govern axon termination in touch receptor neurons. Findings across model organisms, including Caenorhabditis elegans, Drosophila, zebrafish and mice, have revealed that complex signaling is required for termination with conserved principles and players beginning to surface. A key emerging theme is that axon termination is mediated by complex signaling networks that include ubiquitin ligase signaling hubs, kinase cascades, transcription factors, guidance/adhesion receptors and growth factors. Here, we begin a discussion about how these signaling networks could represent termination codes that trigger cessation of axon outgrowth in different species and types of mechanosensory neurons.
Collapse
Affiliation(s)
- Muriel Desbois
- School of Life Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98101, USA
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98101, USA
| |
Collapse
|
3
|
Rajan S, Yoon J, Wu H, Srapyan S, Baskar R, Ahmed G, Yang T, Grintsevich EE, Reisler E, Terman JR. Disassembly of bundled F-actin and cellular remodeling via an interplay of Mical, cofilin, and F-actin crosslinkers. Proc Natl Acad Sci U S A 2023; 120:e2309955120. [PMID: 37725655 PMCID: PMC10523612 DOI: 10.1073/pnas.2309955120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023] Open
Abstract
Cellular form and function are controlled by the assembly and stability of actin cytoskeletal structures-but disassembling/pruning these structures is equally essential for the plasticity and remodeling that underlie behavioral adaptations. Importantly, the mechanisms of actin assembly have been well-defined-including that it is driven by actin's polymerization into filaments (F-actin) and then often bundling by crosslinking proteins into stable higher-order structures. In contrast, it remains less clear how these stable bundled F-actin structures are rapidly disassembled. We now uncover mechanisms that rapidly and extensively disassemble bundled F-actin. Using biochemical, structural, and imaging assays with purified proteins, we show that F-actin bundled with one of the most prominent crosslinkers, fascin, is extensively disassembled by Mical, the F-actin disassembly enzyme. Furthermore, the product of this Mical effect, Mical-oxidized actin, is poorly bundled by fascin, thereby further amplifying Mical's disassembly effects on bundled F-actin. Moreover, another critical F-actin regulator, cofilin, also affects fascin-bundled filaments, but we find herein that it synergizes with Mical to dramatically amplify its disassembly of bundled F-actin compared to the sum of their individual effects. Genetic and high-resolution cellular assays reveal that Mical also counteracts crosslinking proteins/bundled F-actin in vivo to control cellular extension, axon guidance, and Semaphorin/Plexin cell-cell repulsion. Yet, our results also support the idea that fascin-bundling serves to dampen Mical's F-actin disassembly in vitro and in vivo-and that physiologically relevant cellular remodeling requires a fine-tuned interplay between the factors that build bundled F-actin networks and those that disassemble them.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA90095
| | - Jimok Yoon
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Heng Wu
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Sargis Srapyan
- Department of Chemistry and Biochemistry, California State University, Long Beach, CA90840
| | - Raju Baskar
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Giasuddin Ahmed
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Taehong Yang
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Elena E. Grintsevich
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, California State University, Long Beach, CA90840
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA90095
- Molecular Biology Institute, University of California, Los Angeles, CA90095
| | - Jonathan R. Terman
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
4
|
Chen CY, Chao YM, Cho CC, Chen CS, Lin WY, Chen YH, Cassar M, Lu CS, Yang JL, Chan JYH, Juo SHH. Cerebral Semaphorin3D is a novel risk factor for age-associated cognitive impairment. Cell Commun Signal 2023; 21:140. [PMID: 37316917 DOI: 10.1186/s12964-023-01158-5] [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: 02/04/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND We previously reported that miR-195 exerts neuroprotection by inhibiting Sema3A and cerebral miR-195 levels decreased with age, both of which urged us to explore the role of miR-195 and miR-195-regulated Sema3 family members in age-associated dementia. METHODS miR-195a KO mice were used to assess the effect of miR-195 on aging and cognitive functions. Sema3D was predicted as a miR-195 target by TargetScan and then verified by luciferase reporter assay, while effects of Sema3D and miR-195 on neural senescence were assessed by beta-galactosidase and dendritic spine density. Cerebral Sema3D was over-expressed by lentivirus and suppressed by si-RNA, and effects of over-expression of Sema3D and knockdown of miR-195 on cognitive functions were assessed by Morris Water Maze, Y-maze, and open field test. The effect of Sema3D on lifespan was assessed in Drosophila. Sema3D inhibitor was developed using homology modeling and virtual screening. One-way and two-way repeated measures ANOVA were applied to assess longitudinal data on mouse cognitive tests. RESULTS Cognitive impairment and reduced density of dendritic spine were observed in miR-195a knockout mice. Sema3D was identified to be a direct target of miR-195 and a possible contributor to age-associated neurodegeneration as Sema3D levels showed age-dependent increase in rodent brains. Injection of Sema3D-expressing lentivirus caused significant memory deficits while silencing hippocampal Sema3D improved cognition. Repeated injections of Sema3D-expressing lentivirus to elevate cerebral Sema3D for 10 weeks revealed a time-dependent decline of working memory. More importantly, analysis of the data on the Gene Expression Omnibus database showed that Sema3D levels were significantly higher in dementia patients than normal controls (p < 0.001). Over-expression of homolog Sema3D gene in the nervous system of Drosophila reduced locomotor activity and lifespan by 25%. Mechanistically, Sema3D might reduce stemness and number of neural stem cells and potentially disrupt neuronal autophagy. Rapamycin restored density of dendritic spines in the hippocampus from mice injected with Sema3D lentivirus. Our novel small molecule increased viability of Sema3D-treated neurons and might improve autophagy efficiency, which suggested Sema3D could be a potential drug target. Video Abstract CONCLUSION: Our results highlight the importance of Sema3D in age-associated dementia. Sema3D could be a novel drug target for dementia treatment.
Collapse
Affiliation(s)
- Chien-Yuan Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yung-Mei Chao
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ching-Chang Cho
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Sheng Chen
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Psychiatry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Yong Lin
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
- Brain Diseases Research Center, China Medical University, Taichung, Taiwan
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Marlène Cassar
- Formation and Regulation of Neuronal Connectivity Research Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Institut du Cerveau Et de La Moelle Epinière (ICM)-Sorbonne, UniversitéInserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| | - Cecilia S Lu
- Formation and Regulation of Neuronal Connectivity Research Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Jenq-Lin Yang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Julie Y H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Suh-Hang H Juo
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Drug Development Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
5
|
Hai R, You Q, Wu F, Qiu G, Yang Q, Shu L, Xie L, Zhou X. Semaphorin 3D inhibits proliferation and migration of papillary thyroid carcinoma by regulating MAPK/ERK signaling pathway. Mol Biol Rep 2022; 49:3793-3802. [PMID: 35190928 DOI: 10.1007/s11033-022-07220-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/01/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Semaphorin 3D (SEMA3D) plays an important role in the occurrence and development of multifarious cancers. However, the relationship between SEMA3D and papillary thyroid carcinoma (PTC) remains unclear. This study aimed to investigate the functions and mechanism of SEMA3D in papillary thyroid carcinoma (PTC). METHODS The expression of SEMA3D in PTC tissues and cell lines was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Western blotting and immunohistochemistry (IHC) were used to detect the expression of the related proteins. CCK-8 and colony formation assays and Transwell assays were used to evaluate cell proliferation and migration, respectively. A xenograft model was induced to further verify the effect of SEMA3D in vivo. RESULTS In this study, we found that SEMA3D was downregulated in PTC tissues and PTC cell lines (TPC-1 and BCPAP). The expression level of SEMA3D was significantly related to age (P < 0.01), extrathyroidal extension (P < 0.01), TNM stage (P < 0.01) and lymph node metastasis (P < 0.01). In vitro experiments showed that overexpression of SEMA3D inhibited the proliferation and migration of TPC-1 and BCPAP cells and that upregulated SEMA3D inhibited the phosphorylation of ERK and the expression of the phenotype-related proteins PCNA and MMP2. In addition, SEMA3D overexpression inhibited tumour growth in vivo. CONCLUSION In this study, we found that SEMA3D is significantly downregulated in PTC tissues. SEMA3D inhibits the proliferation and migration of PTC cells and suppresses tumour growth in vivo, possibly partially through the MAPK/ERK signalling pathway, suggesting that SEMA3D may be a reliable molecular marker for the diagnosis and treatment of PTC.
Collapse
Affiliation(s)
- Rui Hai
- Department of Breast, Thyroid and Vessel Surgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qian You
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Fei Wu
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Guochun Qiu
- Department of Breast, Thyroid and Vessel Surgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qian Yang
- Department of Oncology, The Leshan People's Hospital, Leshan, 614000, China
| | - Liang Shu
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Linjun Xie
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xiangyu Zhou
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
6
|
Romhányi D, Szabó K, Kemény L, Sebestyén E, Groma G. Transcriptional Analysis-Based Alterations Affecting Neuritogenesis of the Peripheral Nervous System in Psoriasis. Life (Basel) 2022; 12:111. [PMID: 35054504 PMCID: PMC8778302 DOI: 10.3390/life12010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 11/17/2022] Open
Abstract
An increasing amount of evidence indicates the critical role of the cutaneous nervous system in the initiation and maintenance of psoriatic skin lesions by neurogenic inflammation. However, molecular mechanisms affecting cutaneous neurons are largely uncharacterized. Therefore, we reanalyzed a psoriatic RNA sequencing dataset from published transcriptome experiments of nearly 300 individuals. Using the Ingenuity Pathway Analysis software, we associated several hundreds of differentially expressed transcripts (DETs) to nervous system development and functions. Since neuronal projections were previously reported to be affected in psoriasis, we performed an in-depth analysis of neurite formation-related process. Our in silico analysis suggests that SEMA-PLXN and ROBO-DCC-UNC5 regulating axonal growth and repulsion are differentially affected in non-lesional and lesional skin samples. We identified opposing expressional alterations in secreted ligands for axonal guidance signaling (RTN4/NOGOA, NTNs, SEMAs, SLITs) and non-conventional axon guidance regulating ligands, including WNT5A and their receptors, modulating axon formation. These differences in neuritogenesis may explain the abnormal cutaneous nerve filament formation described in psoriatic skin. The processes also influence T-cell activation and infiltration, thus highlighting an additional angle of the crosstalk between the cutaneous nervous system and the immune responses in psoriasis pathogenesis, in addition to the known neurogenic pro-inflammatory mediators.
Collapse
Affiliation(s)
- Dóra Romhányi
- Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (D.R.); (K.S.); (L.K.)
| | - Kornélia Szabó
- Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (D.R.); (K.S.); (L.K.)
- Hungarian Centre of Excellence for Molecular Medicine-University of Szeged Skin Research Group (HCEMM-USZ Skin Research Group), University of Szeged, H-6720 Szeged, Hungary
- Eötvös Loránd Research Network, MTA-SZTE Dermatological Research Group, H-6720 Szeged, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (D.R.); (K.S.); (L.K.)
- Hungarian Centre of Excellence for Molecular Medicine-University of Szeged Skin Research Group (HCEMM-USZ Skin Research Group), University of Szeged, H-6720 Szeged, Hungary
- Eötvös Loránd Research Network, MTA-SZTE Dermatological Research Group, H-6720 Szeged, Hungary
| | - Endre Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary;
| | - Gergely Groma
- Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (D.R.); (K.S.); (L.K.)
- Eötvös Loránd Research Network, MTA-SZTE Dermatological Research Group, H-6720 Szeged, Hungary
| |
Collapse
|
7
|
Haynes EM, Burnett KH, He J, Jean-Pierre MW, Jarzyna M, Eliceiri KW, Huisken J, Halloran MC. KLC4 shapes axon arbors during development and mediates adult behavior. eLife 2022; 11:74270. [PMID: 36222498 PMCID: PMC9596160 DOI: 10.7554/elife.74270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Development of elaborate and polarized neuronal morphology requires precisely regulated transport of cellular cargos by motor proteins such as kinesin-1. Kinesin-1 has numerous cellular cargos which must be delivered to unique neuronal compartments. The process by which this motor selectively transports and delivers cargo to regulate neuronal morphogenesis is poorly understood, although the cargo-binding kinesin light chain (KLC) subunits contribute to specificity. Our work implicates one such subunit, KLC4, as an essential regulator of axon branching and arborization pattern of sensory neurons during development. Using live imaging approaches in klc4 mutant zebrafish, we show that KLC4 is required for stabilization of nascent axon branches, proper microtubule (MT) dynamics, and endosomal transport. Furthermore, KLC4 is required for proper tiling of peripheral axon arbors: in klc4 mutants, peripheral axons showed abnormal fasciculation, a behavior characteristic of central axons. This result suggests that KLC4 patterns axonal compartments and helps establish molecular differences between central and peripheral axons. Finally, we find that klc4 mutant larva are hypersensitive to touch and adults show anxiety-like behavior in a novel tank test, implicating klc4 as a new gene involved in stress response circuits.
Collapse
Affiliation(s)
- Elizabeth M Haynes
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Center for Quantitative Cell Imaging, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States,Morgridge Institute for ResearchMadisonUnited States
| | - Korri H Burnett
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Jiaye He
- Morgridge Institute for ResearchMadisonUnited States,National Innovation Center for Advanced Medical DevicesShenzenChina
| | - Marcel W Jean-Pierre
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Martin Jarzyna
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Kevin W Eliceiri
- Center for Quantitative Cell Imaging, University of Wisconsin-MadisonMadisonUnited States,Morgridge Institute for ResearchMadisonUnited States
| | - Jan Huisken
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Morgridge Institute for ResearchMadisonUnited States,Department of Biology and Psychology, Georg-August-UniversityGöttingenGermany
| | - Mary C Halloran
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
8
|
Shorey M, Rao K, Stone MC, Mattie FJ, Sagasti A, Rolls MM. Microtubule organization of vertebrate sensory neurons in vivo. Dev Biol 2021; 478:1-12. [PMID: 34147472 DOI: 10.1016/j.ydbio.2021.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 01/30/2023]
Abstract
Dorsal root ganglion (DRG) neurons are the predominant cell type that innervates the vertebrate skin. They are typically described as pseudounipolar cells that have central and peripheral axons branching from a single root exiting the cell body. The peripheral axon travels within a nerve to the skin, where free sensory endings can emerge and branch into an arbor that receives and integrates information. In some immature vertebrates, DRG neurons are preceded by Rohon-Beard (RB) neurons. While the sensory endings of RB and DRG neurons function like dendrites, we use live imaging in zebrafish to show that they have axonal plus-end-out microtubule polarity at all stages of maturity. Moreover, we show both cell types have central and peripheral axons with plus-end-out polarity. Surprisingly, in DRG neurons these emerge separately from the cell body, and most cells never acquire the signature pseudounipolar morphology. Like another recently characterized cell type that has multiple plus-end-out neurites, ganglion cells in Nematostella, RB and DRG neurons maintain a somatic microtubule organizing center even when mature. In summary, we characterize key cellular and subcellular features of vertebrate sensory neurons as a foundation for understanding their function and maintenance.
Collapse
Affiliation(s)
- Matthew Shorey
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Kavitha Rao
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Michelle C Stone
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Floyd J Mattie
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Alvaro Sagasti
- Molecular, Cell and Developmental Biology Department and Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Melissa M Rolls
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
9
|
Holt E, Stanton-Turcotte D, Iulianella A. Development of the Vertebrate Trunk Sensory System: Origins, Specification, Axon Guidance, and Central Connectivity. Neuroscience 2021; 458:229-243. [PMID: 33460728 DOI: 10.1016/j.neuroscience.2020.12.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/09/2020] [Accepted: 12/31/2020] [Indexed: 12/26/2022]
Abstract
Crucial to an animal's movement through their environment and to the maintenance of their homeostatic physiology is the integration of sensory information. This is achieved by axons communicating from organs, muscle spindles and skin that connect to the sensory ganglia composing the peripheral nervous system (PNS), enabling organisms to collect an ever-constant flow of sensations and relay it to the spinal cord. The sensory system carries a wide spectrum of sensory modalities - from sharp pain to cool refreshing touch - traveling from the periphery to the spinal cord via the dorsal root ganglia (DRG). This review covers the origins and development of the DRG and the cells that populate it, and focuses on how sensory connectivity to the spinal cord is achieved by the diverse developmental and molecular processes that control axon guidance in the trunk sensory system. We also describe convergences and differences in sensory neuron formation among different vertebrate species to gain insight into underlying developmental mechanisms.
Collapse
Affiliation(s)
- Emily Holt
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia B3H-4R2, Canada
| | - Danielle Stanton-Turcotte
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia B3H-4R2, Canada
| | - Angelo Iulianella
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia B3H-4R2, Canada.
| |
Collapse
|
10
|
Suter TACS, Jaworski A. Cell migration and axon guidance at the border between central and peripheral nervous system. Science 2020; 365:365/6456/eaaw8231. [PMID: 31467195 DOI: 10.1126/science.aaw8231] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/22/2019] [Indexed: 12/13/2022]
Abstract
The central and peripheral nervous system (CNS and PNS, respectively) are composed of distinct neuronal and glial cell types with specialized functional properties. However, a small number of select cells traverse the CNS-PNS boundary and connect these two major subdivisions of the nervous system. This pattern of segregation and selective connectivity is established during embryonic development, when neurons and glia migrate to their destinations and axons project to their targets. Here, we provide an overview of the cellular and molecular mechanisms that control cell migration and axon guidance at the vertebrate CNS-PNS border. We highlight recent advances on how cell bodies and axons are instructed to either cross or respect this boundary, and present open questions concerning the development and plasticity of the CNS-PNS interface.
Collapse
Affiliation(s)
- Tracey A C S Suter
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA.,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912, USA
| | - Alexander Jaworski
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA. .,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912, USA
| |
Collapse
|
11
|
Liu L, Zhao M, Xie ZG, Liu J, Peng HP, Pei YF, Sun HP, Zhang L. Twelve New Genomic Loci Associated With Bone Mineral Density. Front Endocrinol (Lausanne) 2020; 11:243. [PMID: 32390946 PMCID: PMC7188784 DOI: 10.3389/fendo.2020.00243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/02/2020] [Indexed: 12/17/2022] Open
Abstract
Aiming to identify more genomic loci associated with bone mineral density (BMD), we conducted a joint association analysis of 2 genome-wide association study (GWAS) by the integrative association method multi-trait analysis of GWAS (MTAG). The first one is the single GWAS of estimated heel BMD (eBMD) in the UK biobank (UKB) cohort (N = 426,824), and the second one is the GWAS meta-analysis of total body BMD (TB-BMD) in 66,628 participants from 30 studies. Approximate conditional association analysis was performed in the identified novel loci to identify secondary association signal. Statistical fine-mapping was conducted to prioritize plausible credible risk variants (CRVs). Candidate genes were prioritized based on the analyses of cis- expression quantitative trait locus (cis-eQTL) and cis-protein QTL (cis-pQTL) information as well as the functional category of the SNP. By integrating the information carried in over 490,000 participants, this largest joint analysis of BMD GWAS identified 12 novel genomic loci at the genome-wide significance level (GWS, p = 5.0 × 10-8), nine of which were for eBMD and four were for TB-BMD, explaining an additional 0.11 and 0.23% heritability for the two traits, respectively. These loci include 1p33 (lead SNP rs10493130, peBMD = 3.19 × 10-8), 5q13.2 (rs4703589, peBMD = 4.78 × 10-8), 5q31.3 (rs9324887, pTB-BMD = 1.36 × 10-9), 6p21.32 (rs6905837, peBMD = 3.32 × 10-8), 6q14.1 (rs10806234, peBMD = 2.63 × 10-8), 7q21.11 (rs10806234, pTB-BMD = 3.37 × 10-8), 8q24.12 (rs11995866, peBMD = 6.72 × 10-9), 12p13.31 (rs1639122, peBMD = 4.43 × 10-8), 12p12.1 (rs58489179, peBMD = 4.74 × 10-8), 12q24.23 (rs75499226, peBMD = 1.44 × 10-8), 19q13.31 (rs7255083, pTB-BMD = 2.18 × 10-8) and 22q11.23 (rs13056137, pTB-BMD = 2.54 × 10-8). All lead SNPs in these 12 loci are nominally significant in both original studies as well as consistent in effect direction between them, providing solid evidence of replication. Approximate conditional analysis identified one secondary signal in 5q13.2 (rs11738874, pconditional = 5.06 × 10-9). Statistical fine-mapping analysis prioritized 269 CRVs. A total of 65 candidate genes were prioritized, including those with known biological function to bone development (such as FGF1, COL11A2 and DEPTOR). Our findings provide novel insights into a better understanding of the genetic mechanism underlying bone development as well as candidate genes for future functional investigation.
Collapse
Affiliation(s)
- Lu Liu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Jiangsu, China
- Kunshan Hospital of Traditional Chinese Medicine, Jiangsu, China
| | - Min Zhao
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Jiangsu, China
| | - Zong-Gang Xie
- The Second Affiliated Hospital of Soochow University, Jiangsu, China
| | - Ju Liu
- Kunshan Hospital of Traditional Chinese Medicine, Jiangsu, China
| | - Hui-Ping Peng
- Kunshan Hospital of Traditional Chinese Medicine, Jiangsu, China
| | - Yu-Fang Pei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, China
- *Correspondence: Yu-Fang Pei
| | - Hong-Peng Sun
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Jiangsu, China
- Department of Children Health and Social Medicine, School of Public Health, Medical College of Soochow University, Jiangsu, China
- Hong-Peng Sun
| | - Lei Zhang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Jiangsu, China
- Lei Zhang
| |
Collapse
|
12
|
Freeman L, Wu OC, Sweet J, Cohen M, Smith GA, Miller JP. Facial Sensory Restoration After Trigeminal Sensory Rhizotomy by Collateral Sprouting From the Occipital Nerves. Neurosurgery 2019; 86:E436-E441. [DOI: 10.1093/neuros/nyz306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/18/2019] [Indexed: 01/14/2023] Open
Abstract
Abstract
BACKGROUND AND IMPORTANCE
Lesioning procedures are effective for trigeminal neuralgia (TN), but late pain recurrence associated with sensory recovery is common. We report a case of recurrence of type 1A TN and recovery of facial sensory function after trigeminal rhizotomy associated with collateral sprouting from upper cervical spinal nerves.
CLINICAL PRESENTATION
A 41-yr-old woman presented 2 yr after open left trigeminal sensory rhizotomy for TN with pain-free anesthesia in the entire left trigeminal nerve distribution. Over 18 mo, she developed gradual recovery of facial sensation migrating anteromedially from the occipital region, eventually extending to the midpupillary line across the distribution of all trigeminal nerve branches. She reported recurrence of her triggered lancinating TN pain isolated to the area of recovered sensation with no pain in anesthetic areas. Nerve ultrasound demonstrated enlargement of ipsilateral greater and lesser occipital nerves, and occipital nerve block restored facial anesthesia and resolved her pain, indicating that recovered facial sensation was provided exclusively by the upper cervical spinal nerves. She underwent C2/C3 ganglionectomy, and ganglia were observed to be hypertrophic. Postoperatively, trigeminal anesthesia was restored with complete resolution of pain that persisted at 12-mo follow-up.
CONCLUSION
This is the first documented case of a spinal nerve innervating a cranial dermatome by collateral sprouting after cranial nerve injury. The fact that typical TN pain can occur even when sensation is mediated by spinal nerves suggests that the disorder can be centrally mediated and late failure after lesioning procedures may result from maladaptive reinnervation.
Collapse
Affiliation(s)
- Lindsey Freeman
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Osmond C Wu
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jennifer Sweet
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Mark Cohen
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Gabriel A Smith
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jonathan P Miller
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
13
|
Nazaryan-Petersen L, Oliveira IR, Mehrjouy MM, Mendez JMM, Bak M, Bugge M, Kalscheuer VM, Bache I, Hancks DC, Tommerup N. Multigenic truncation of the semaphorin-plexin pathway by a germline chromothriptic rearrangement associated with Moebius syndrome. Hum Mutat 2019; 40:1057-1062. [PMID: 31033088 DOI: 10.1002/humu.23775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/17/2019] [Accepted: 04/24/2019] [Indexed: 11/07/2022]
Abstract
Moebius syndrome (MBS) is a congenital disorder caused by paralysis of the facial and abducens nerves. Although a number of candidate genes have been suspected, so far only mutations in PLXND1 and REV3L are confirmed to cause MBS. Here, we fine mapped the breakpoints of a complex chromosomal rearrangement (CCR) 46,XY,t(7;8;11;13) in a patient with MBS, which revealed 41 clustered breakpoints with typical hallmarks of chromothripsis. Among 12 truncated protein-coding genes, SEMA3A is known to bind to the MBS-associated PLXND1. Intriguingly, the CCR also truncated PIK3CG, which in silico interacts with REVL3 encoded by the other known MBS-gene REV3L, and with the SEMA3A/PLXND1 complex via FLT1. Additional studies of other complex rearrangements may reveal whether the multiple breakpoints in germline chromothripsis may predispose to complex multigenic disorders.
Collapse
Affiliation(s)
- Lusine Nazaryan-Petersen
- Wilhelm Johannsen Center for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Inês R Oliveira
- Wilhelm Johannsen Center for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Regulation and Evaluation of Medicines and Health products, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Mana M Mehrjouy
- Wilhelm Johannsen Center for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Juan M M Mendez
- Wilhelm Johannsen Center for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mads Bak
- Wilhelm Johannsen Center for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Merete Bugge
- Wilhelm Johannsen Center for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Vera M Kalscheuer
- Research Group Development and Disease, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Iben Bache
- Wilhelm Johannsen Center for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Dustin C Hancks
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Niels Tommerup
- Wilhelm Johannsen Center for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Wang S, Su D, Li J, Li D, Wan H, Schumacher M, Liu S. Donor nerve axotomy and axonal regeneration after end-to-side neurorrhaphy in a rodent model. J Neurosurg 2019; 130:197-206. [PMID: 29451448 DOI: 10.3171/2017.8.jns17739] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/08/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE In this study, the authors used a surgical model of end-to-side neurorrhaphy between a nerve graft and a donor tibial nerve in adult rats to investigate the optimal conditions for axonal regeneration induced by the donor nerve. They also assessed the importance of a more favorable pathway using a predegenerated nerve graft to attract regenerating axons to regrow into the graft and then directing and improving their growth toward the target in comparison with results obtained with a fresh nerve graft. METHODS End-to-side neurorrhaphy was performed between a nerve graft and a donor tibial nerve. The nerve graft was obtained from the left tibial nerve, which was either freshly removed or predegenerated 1 week prior to neurorrhaphy. The donor right tibial nerve was injured by epineurium removal alone, injured by epineurium removal with cross section of 20% or 50% of the total axons at the coaptation site, or left intact. The animals were followed postoperatively for a 6-week period, and outcomes were evaluated by optical microscopy and retrograde labeling to detect the regenerated primary sensory neurons located in the lumbar dorsal root ganglia and spinal motor neurons located in the lumbar spinal ventral horn. RESULTS At the end of the follow-up period, no regenerating axons were observed in the nerve grafts when the donor nerve was left intact, and very few axons were detected when the donor nerve was injured by epineurium removal alone. However, numerous regenerating axons appeared in the grafts when the donor nerve was axotomized, and the greatest number was achieved with a 50% cross section axotomized nerve. In the rats with a 50% cross section of the donor nerve, better nerve-like morphology of the grafts was observed, without connective adhesions. When a predegenerated nerve graft was used, more regenerating axons were attracted and elongated with a more regular shape and improved myelination. CONCLUSIONS Axonal regrowth into a nerve graft depends on axotomy of the donor nerve after end-to-side neurorrhaphy. More efficient attraction and an improved structure of the regenerating axons were achieved when a predegenerated nerve graft was used. Furthermore, a nerve graft may require a certain number of regenerating axons to maintain a nerve-like morphology.
Collapse
Affiliation(s)
- Shiwei Wang
- 2U 1195, INSERM, Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France; and
| | - Diya Su
- 1Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jing Li
- 1Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Dezhi Li
- 3Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong Wan
- 1Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Michael Schumacher
- 2U 1195, INSERM, Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France; and
| | - Song Liu
- 1Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- 2U 1195, INSERM, Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France; and
| |
Collapse
|
15
|
Liu CF, Angelozzi M, Haseeb A, Lefebvre V. SOX9 is dispensable for the initiation of epigenetic remodeling and the activation of marker genes at the onset of chondrogenesis. Development 2018; 145:dev164459. [PMID: 30021842 PMCID: PMC6078338 DOI: 10.1242/dev.164459] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022]
Abstract
SOX9 controls cell lineage fate and differentiation in major biological processes. It is known as a potent transcriptional activator of differentiation-specific genes, but its earliest targets and its contribution to priming chromatin for gene activation remain unknown. Here, we address this knowledge gap using chondrogenesis as a model system. By profiling the whole transcriptome and the whole epigenome of wild-type and Sox9-deficient mouse embryo limb buds, we uncover multiple structural and regulatory genes, including Fam101a, Myh14, Sema3c and Sema3d, as specific markers of precartilaginous condensation, and we provide evidence of their direct transactivation by SOX9. Intriguingly, we find that SOX9 helps remove epigenetic signatures of transcriptional repression and establish active-promoter and active-enhancer marks at precartilage- and cartilage-specific loci, but is not absolutely required to initiate these changes and activate transcription. Altogether, these findings widen our current knowledge of SOX9 targets in early chondrogenesis and call for new studies to identify the pioneer and transactivating factors that act upstream of or along with SOX9 to prompt chromatin remodeling and specific gene activation at the onset of chondrogenesis and other processes.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Marco Angelozzi
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Abdul Haseeb
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Véronique Lefebvre
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| |
Collapse
|
16
|
Gurung S, Asante E, Hummel D, Williams A, Feldman-Schultz O, Halloran MC, Sittaramane V, Chandrasekhar A. Distinct roles for the cell adhesion molecule Contactin2 in the development and function of neural circuits in zebrafish. Mech Dev 2018; 152:1-12. [PMID: 29777776 DOI: 10.1016/j.mod.2018.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/02/2018] [Accepted: 05/09/2018] [Indexed: 01/17/2023]
Abstract
Contactin2 (Cntn2)/Transient Axonal Glycoprotein 1 (Tag1), a neural cell adhesion molecule, has established roles in neuronal migration and axon fasciculation in chick and mouse. In zebrafish, antisense morpholino-based studies have indicated roles for cntn2 in the migration of facial branchiomotor (FBM) neurons, the guidance of the axons of the nucleus of the medial longitudinal fascicle (nucMLF), and the outgrowth of Rohon-Beard (RB) central axons. To study functions of Cntn2 in later stages of neuronal development, we generated cntn2 mutant zebrafish using CRISPR-Cas9. Using a null mutant allele, we detected genetic interactions between cntn2 and the planar cell polarity gene vangl2, as shown previously with cntn2 morphants, demonstrating a function for cntn2 during FBM neuron migration in a sensitized background of reduced planar cell polarity signaling. In addition, maternal-zygotic (MZ) cntn2 mutant larvae exhibited aberrant touch responses and swimming, suggestive of defects in sensorimotor circuits, consistent with studies in mice. However, the nucMLF axon convergence, FBM neuron migration, and RB outgrowth defects seen in morphants were not seen in the mutants, and we show here that they are likely off-target effects of morpholinos. However, MLF axons exhibited local defasciculation in MZcntn2 mutants, consistent with a role for Cntn2 in axon fasciculation. These data demonstrate distinct roles for zebrafish cntn2 in neuronal migration and axon fasciculation, and in the function of sensorimotor circuits.
Collapse
Affiliation(s)
- Suman Gurung
- Division of Biological Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Emilia Asante
- Division of Biological Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Devynn Hummel
- Division of Biological Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Ashley Williams
- Department of Biology, Georgia Southern University, Statesboro, GA 30458, USA
| | - Oren Feldman-Schultz
- Department of Integrative Biology, Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Mary C Halloran
- Department of Integrative Biology, Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Vinoth Sittaramane
- Department of Biology, Georgia Southern University, Statesboro, GA 30458, USA
| | - Anand Chandrasekhar
- Division of Biological Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
17
|
Nozawa K, Lin Y, Kubodera R, Shimizu Y, Tanaka H, Ohshima T. Zebrafish Mecp2 is required for proper axonal elongation of motor neurons and synapse formation. Dev Neurobiol 2017; 77:1101-1113. [PMID: 28371371 DOI: 10.1002/dneu.22498] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/08/2017] [Accepted: 03/23/2017] [Indexed: 12/13/2022]
Abstract
Rett syndrome is a severe neurodevelopmental disorder. It is caused by a mutation in methyl-CpG binding protein 2 (MecP2), a transcriptional regulator that recruits protein complexes involved in histone modification and chromatin remodeling. However, the role of Mecp2 in Rett syndrome remains unclear. In this study, we investigated the function of Mecp2 in neuronal development using zebrafish embryos. Mecp2 expression was detected ubiquitously in the central nervous system and muscles at 28 h postfertilization (hpf). We injected an antisense morpholino oligonucleotide (AMO) to induce Mecp2 knockdown phenotype. In mecp2 morphants (embryos with Mecp2 knockdown by AMO) at 28 and 72 hpf, we found an increase in abnormal axonal branches of caudal primary motor neurons and a decrease in motor activity. In mecp2 morphants at 24 hpf, we observed an increase in the expression of an mecp2 downstream candidate gene, brain derived neurotrophic factor (bdnf). In mecp2 morphants at 72 hpf, the presynaptic area stained by an anti-SV2 antibody was increased at the neuromuscular junction (NMJ). Interestingly, the size of SV2-positive presynaptic area at the NMJ was also increased following bdnf mRNA injection, while it was normalized in a double knockdown of mecp2 and bdnf. These results imply that Mecp2 is an important functional regulator of bdnf gene expression during neural circuit formation in zebrafish embryo. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1101-1113, 2017.
Collapse
Affiliation(s)
- Keisuke Nozawa
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480, Japan
| | - Yanbin Lin
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480, Japan
| | - Ryota Kubodera
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480, Japan
| | - Yuki Shimizu
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480, Japan
| | - Hideomi Tanaka
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480, Japan
| | - Toshio Ohshima
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480, Japan
| |
Collapse
|
18
|
Lee TJ, Lee JW, Haynes EM, Eliceiri KW, Halloran MC. The Kinesin Adaptor Calsyntenin-1 Organizes Microtubule Polarity and Regulates Dynamics during Sensory Axon Arbor Development. Front Cell Neurosci 2017; 11:107. [PMID: 28473757 PMCID: PMC5397401 DOI: 10.3389/fncel.2017.00107] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/29/2017] [Indexed: 11/30/2022] Open
Abstract
Axon growth and branching, and development of neuronal polarity are critically dependent on proper organization and dynamics of the microtubule (MT) cytoskeleton. MTs must organize with correct polarity for delivery of diverse cargos to appropriate subcellular locations, yet the molecular mechanisms regulating MT polarity remain poorly understood. Moreover, how an actively branching axon reorganizes MTs to direct their plus ends distally at branch points is unknown. We used high-speed, in vivo imaging of polymerizing MT plus ends to characterize MT dynamics in developing sensory axon arbors in zebrafish embryos. We find that axonal MTs are highly dynamic throughout development, and that the peripheral and central axons of sensory neurons show differences in MT behaviors. Furthermore, we show that Calsyntenin-1 (Clstn-1), a kinesin adaptor required for sensory axon branching, also regulates MT polarity in developing axon arbors. In wild type neurons the vast majority of MTs are directed in the correct plus-end-distal orientation from early stages of development. Loss of Clstn-1 causes an increase in MTs polymerizing in the retrograde direction. These misoriented MTs most often are found near growth cones and branch points, suggesting Clstn-1 is particularly important for organizing MT polarity at these locations. Together, our results suggest that Clstn-1, in addition to regulating kinesin-mediated cargo transport, also organizes the underlying MT highway during axon arbor development.
Collapse
Affiliation(s)
- Tristan J Lee
- Department of Zoology, University of Wisconsin-MadisonMadison, WI, USA.,Department of Neuroscience, University of Wisconsin-MadisonMadison, WI, USA.,Neuroscience Training Program, University of Wisconsin-MadisonMadison, WI, USA
| | - Jacob W Lee
- Department of Zoology, University of Wisconsin-MadisonMadison, WI, USA.,Department of Neuroscience, University of Wisconsin-MadisonMadison, WI, USA
| | - Elizabeth M Haynes
- Department of Zoology, University of Wisconsin-MadisonMadison, WI, USA.,Department of Neuroscience, University of Wisconsin-MadisonMadison, WI, USA.,Laboratory for Optical and Computational Instrumentation, University of Wisconsin-MadisonMadison, WI, USA
| | - Kevin W Eliceiri
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-MadisonMadison, WI, USA
| | - Mary C Halloran
- Department of Zoology, University of Wisconsin-MadisonMadison, WI, USA.,Department of Neuroscience, University of Wisconsin-MadisonMadison, WI, USA.,Neuroscience Training Program, University of Wisconsin-MadisonMadison, WI, USA
| |
Collapse
|
19
|
Wang Z, Ding M, Qian N, Song B, Yu J, Tang J, Wang J. Decreased expression of semaphorin 3D is associated with genesis and development in colorectal cancer. World J Surg Oncol 2017; 15:67. [PMID: 28320475 PMCID: PMC5359842 DOI: 10.1186/s12957-017-1128-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 03/06/2017] [Indexed: 12/17/2022] Open
Abstract
Background Semaphorin 3D (SEMA3D) plays important roles in the genesis and progress of many cancers. However, the relationship between SEMA3D and colorectal cancer (CRC) remains unknown. The aim of this study was to investigate whether SEMA3D can be used as a predictive marker for the diagnosis, metastasis, and prognosis of CRC by assessing the expression of SEMA3D in the tissues and serum of CRC patients. Methods Real-time quantitative polymerase chain reaction (qPCR) was used to measure the expression of SEMA3D mRNA in 100 CRC tissues and matched normal tissues. qPCR was also used to detect the expression of SEMA3D mRNA in the CRC cell line RKO. RKO cells were transfected with SEMA3D small-interring RNA (siRNA) to interfere with endogenous SEMA3D. The migratory ability of control and SEMA3D siRNA-transfected RKO cells was determined by transwell assays. Enzyme-linked immunosorbent assay (ELISA) was utilized to detect the levels of SEMA3D in the serum of 80 CRC patients and 100 normal healthy controls. The expression of SEMA3D in 215 CRC tissues was assessed using immunohistochemistry (IHC). Then, statistical analyses were adopted to assess SEMA3D protein levels and clinical pathological characteristics. Results The mRNA expression of SEMA3D was significantly lower in CRC tissues than in paired normal tissues (t = 5.027, P < 0.0001). Compared with normal healthy controls, the serum levels of SEMA3D were decreased significantly in CRC patients (t = 3.656, P = 0.0003). The expression of SEMA3D protein was linked to lymph node metastasis, and low expression led to lymph node metastasis (χ2 = 8.415, P = 0.004). The expression of SEMA3D in CRC tissues was a favorable prognostic factor. Patients with a higher expression of SEMA3D experienced longer survival (P = 0.002, log-rank [Mantel-Cox]; Kaplan-Meier). In addition, multivariate Cox’s proportional hazard model revealed that SEMA3D is an independent prognostic marker (hazard ratio [HR] 1.818, 95% CI 1.063–3.110, P = 0.029). Moreover, transwell assays showed that knocking down SEMA3D significantly increased RKO cell migration (t = 9.268, P = 0.0008). Conclusions SEMA3D might function as a tumor suppressor during the formation and development of CRC. SEMA3D might become a predictive marker for the diagnosis, metastasis, and prognosis of CRC and provide a novel target for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Pathology, The First Hospital of Jiaxing, Zhejiang, People's Republic of China
| | - Meiman Ding
- The Criminal Investigation Detachment of Jiaxing Public Security Bureau, Zhejiang, People's Republic of China
| | - Naiying Qian
- Department of Pathology, The First Hospital of Jiaxing, Zhejiang, People's Republic of China
| | - Beifeng Song
- Department of Pathology, The First Hospital of Jiaxing, Zhejiang, People's Republic of China
| | - Jiayin Yu
- Department of Pathology, The First Hospital of Jiaxing, Zhejiang, People's Republic of China
| | - Jinlong Tang
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jingyu Wang
- Department of Pathology, The First Hospital of Jiaxing, Zhejiang, People's Republic of China.
| |
Collapse
|
20
|
Gennarini G, Bizzoca A, Picocci S, Puzzo D, Corsi P, Furley AJW. The role of Gpi-anchored axonal glycoproteins in neural development and neurological disorders. Mol Cell Neurosci 2016; 81:49-63. [PMID: 27871938 DOI: 10.1016/j.mcn.2016.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 01/06/2023] Open
Abstract
This review article focuses on the Contactin (CNTN) subset of the Immunoglobulin supergene family (IgC2/FNIII molecules), whose components share structural properties (the association of Immunoglobulin type C2 with Fibronectin type III domains), as well as a general role in cell contact formation and axonal growth control. IgC2/FNIII molecules include 6 highly related components (CNTN 1-6), associated with the cell membrane via a Glycosyl Phosphatidyl Inositol (GPI)-containing lipid tail. Contactin 1 and Contactin 2 share ~50 (49.38)% identity at the aminoacid level. They are components of the cell surface, from which they may be released in soluble forms. They bind heterophilically to multiple partners in cis and in trans, including members of the related L1CAM family and of the Neurexin family Contactin-associated proteins (CNTNAPs or Casprs). Such interactions are important for organising the neuronal membrane, as well as for modulating the growth and pathfinding of axon tracts. In addition, they also mediate the functional maturation of axons by promoting their interactions with myelinating cells at the nodal, paranodal and juxtaparanodal regions. Such interactions also mediate differential ionic channels (both Na+ and K+) distribution, which is of critical relevance in the generation of the peak-shaped action potential. Indeed, thanks to their interactions with Ankyrin G, Na+ channels map within the nodal regions, where they drive axonal depolarization. However, no ionic channels are found in the flanking Contactin1-containing paranodal regions, where CNTN1 interactions with Caspr1 and with the Ig superfamily component Neurofascin 155 in cis and in trans, respectively, build a molecular barrier between the node and the juxtaparanode. In this region K+ channels are clustered, depending upon molecular interactions with Contactin 2 and with Caspr2. In addition to these functions, the Contactins appear to have also a role in degenerative and inflammatory disorders: indeed Contactin 2 is involved in neurodegenerative disorders with a special reference to the Alzheimer disease, given its ability to work as a ligand of the Alzheimer Precursor Protein (APP), which results in increased Alzheimer Intracellular Domain (AICD) release in a γ-secretase-dependent manner. On the other hand Contactin 1 drives Notch signalling activation via the Hes pathway, which could be consistent with its ability to modulate neuroinflammation events, and with the possibility that Contactin 1-dependent interactions may participate to the pathogenesis of the Multiple Sclerosis and of other inflammatory disorders.
Collapse
Affiliation(s)
- Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy.
| | - Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Sabrina Picocci
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Patrizia Corsi
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Andrew J W Furley
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2NT, UK
| |
Collapse
|
21
|
Zwarts L, Goossens T, Clements J, Kang YY, Callaerts P. Axon Branch-Specific Semaphorin-1a Signaling in Drosophila Mushroom Body Development. Front Cell Neurosci 2016; 10:210. [PMID: 27656129 PMCID: PMC5011136 DOI: 10.3389/fncel.2016.00210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/23/2016] [Indexed: 11/25/2022] Open
Abstract
Correct wiring of the mushroom body (MB) neuropil in the Drosophila brain involves appropriate positioning of different axonal lobes, as well as the sister branches that develop from individual axons. This positioning requires the integration of various guidance cues provided by different cell types, which help the axons find their final positions within the neuropil. Semaphorins are well-known for their conserved roles in neuronal development and axon guidance. We investigated the role of Sema-1a in MB development more closely. We show that Sema-1a is expressed in the MBs as well as surrounding structures, including the glial transient interhemispheric fibrous ring, throughout development. By loss- and gain-of-function experiments, we show that the MB axons display lobe and sister branch-specific Sema-1a signaling, which controls different aspects of axon outgrowth and guidance. Furthermore, we demonstrate that these effects are modulated by the integration of MB intrinsic and extrinsic Sema-1a signaling pathways involving PlexA and PlexB. Finally, we also show a role for neuronal- glial interaction in Sema-1a dependent β-lobe outgrowth.
Collapse
Affiliation(s)
- Liesbeth Zwarts
- Laboratory of Behavioral and Developmental Genetics, Department of Human Genetics, KU Leuven, LeuvenBelgium; Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, LeuvenBelgium
| | - Tim Goossens
- Laboratory of Behavioral and Developmental Genetics, Department of Human Genetics, KU Leuven, LeuvenBelgium; Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, LeuvenBelgium
| | - Jason Clements
- Laboratory of Behavioral and Developmental Genetics, Department of Human Genetics, KU Leuven, LeuvenBelgium; Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, LeuvenBelgium
| | - Yuan Y Kang
- Department of Biology and Biochemistry, University of Houston, Houston, TX USA
| | - Patrick Callaerts
- Laboratory of Behavioral and Developmental Genetics, Department of Human Genetics, KU Leuven, LeuvenBelgium; Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, LeuvenBelgium
| |
Collapse
|
22
|
Frei JA, Stoeckli ET. SynCAMs - From axon guidance to neurodevelopmental disorders. Mol Cell Neurosci 2016; 81:41-48. [PMID: 27594578 DOI: 10.1016/j.mcn.2016.08.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 08/28/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022] Open
Abstract
Many cell adhesion molecules are located at synapses but only few of them can be considered synaptic cell adhesion molecules in the strict sense. Besides the Neurexins and Neuroligins, the LRRTMs (leucine rich repeat transmembrane proteins) and the SynCAMs/CADMs can induce synapse formation when expressed in non-neuronal cells and therefore are true synaptic cell adhesion molecules. SynCAMs (synaptic cell adhesion molecules) are a subfamily of the immunoglobulin superfamily of cell adhesion molecules. As suggested by their name, they were first identified as cell adhesion molecules at the synapse which were sufficient to trigger synapse formation. They also contribute to myelination by mediating axon-glia cell contacts. More recently, their role in earlier stages of neural circuit formation was demonstrated, as they also guide axons both in the peripheral and in the central nervous system. Mutations in SynCAM genes were found in patients diagnosed with autism spectrum disorders. The diverse functions of SynCAMs during development suggest that neurodevelopmental disorders are not only due to defects in synaptic plasticity. Rather, early steps of neural circuit formation are likely to contribute.
Collapse
Affiliation(s)
- Jeannine A Frei
- Hussman Institute for Autism, 801 W Baltimore Street, Baltimore, MD 20201, United States
| | - Esther T Stoeckli
- Dept of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
23
|
Dascenco D, Erfurth ML, Izadifar A, Song M, Sachse S, Bortnick R, Urwyler O, Petrovic M, Ayaz D, He H, Kise Y, Thomas F, Kidd T, Schmucker D. Slit and Receptor Tyrosine Phosphatase 69D Confer Spatial Specificity to Axon Branching via Dscam1. Cell 2015; 162:1140-54. [PMID: 26317474 PMCID: PMC4699798 DOI: 10.1016/j.cell.2015.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 06/30/2015] [Accepted: 07/10/2015] [Indexed: 11/26/2022]
Abstract
Axonal branching contributes substantially to neuronal circuit complexity. Studies in Drosophila have shown that loss of Dscam1 receptor diversity can fully block axon branching in mechanosensory neurons. Here we report that cell-autonomous loss of the receptor tyrosine phosphatase 69D (RPTP69D) and loss of midline-localized Slit inhibit formation of specific axon collaterals through modulation of Dscam1 activity. Genetic and biochemical data support a model in which direct binding of Slit to Dscam1 enhances the interaction of Dscam1 with RPTP69D, stimulating Dscam1 dephosphorylation. Single-growth-cone imaging reveals that Slit/RPTP69D are not required for general branch initiation but instead promote the extension of specific axon collaterals. Hence, although regulation of intrinsic Dscam1-Dscam1 isoform interactions is essential for formation of all mechanosensory-axon branches, the local ligand-induced alterations of Dscam1 phosphorylation in distinct growth-cone compartments enable the spatial specificity of axon collateral formation.
Collapse
Affiliation(s)
- Dan Dascenco
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Maria-Luise Erfurth
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium; Institute of Biochemistry, Christian-Albrechts-University of Kiel, Olshausenstr. 40, 24098 Kiel, Germany
| | - Azadeh Izadifar
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Minmin Song
- Biology/MS 314, University of Nevada, Reno, NV 89557, USA
| | - Sonja Sachse
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Biology, Chemistry & Pharmacy, Free University Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Rachel Bortnick
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Olivier Urwyler
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Milan Petrovic
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Derya Ayaz
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Haihuai He
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Yoshiaki Kise
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Franziska Thomas
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Thomas Kidd
- Biology/MS 314, University of Nevada, Reno, NV 89557, USA
| | - Dietmar Schmucker
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
24
|
Kostadinov D, Sanes JR. Protocadherin-dependent dendritic self-avoidance regulates neural connectivity and circuit function. eLife 2015; 4. [PMID: 26140686 PMCID: PMC4548410 DOI: 10.7554/elife.08964] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 07/02/2015] [Indexed: 12/30/2022] Open
Abstract
Dendritic and axonal arbors of many neuronal types exhibit self-avoidance, in which branches repel each other. In some cases, these neurites interact with those of neighboring neurons, a phenomenon called self/non-self discrimination. The functional roles of these processes remain unknown. In this study, we used retinal starburst amacrine cells (SACs), critical components of a direction-selective circuit, to address this issue. In SACs, both processes are mediated by the gamma-protocadherins (Pcdhgs), a family of 22 recognition molecules. We manipulated Pcdhg expression in SACs and recorded from them and their targets, direction-selective ganglion cells (DSGCs). SACs form autapses when self-avoidance is disrupted and fail to form connections with other SACs when self/non-self discrimination is perturbed. Pcdhgs are also required to prune connections between closely spaced SACs. These alterations degrade the direction selectivity of DSGCs. Thus, self-avoidance, self/non-self discrimination, and synapse elimination are essential for proper function of a circuit that computes directional motion. DOI:http://dx.doi.org/10.7554/eLife.08964.001 Nerve cells (or neurons) connect to one another to form circuits that control the animal's behavior. Typically, each neuron receives signals from other cells via branch-like structures called dendrites. Each specific type of neuron has a characteristic pattern of branched dendrites, which is different from the pattern of other types of neuron. Therefore, it is reasonable to imagine that the shape of these branches can influence how the neuron works; however, this idea has rarely been tested experimentally. Different processes are known to act together to control the pattern of the branched dendrites. For example, dendrites in some neurons avoid other dendrites from the same neuron. This phenomenon is referred to as ‘self-avoidance’. In some of these cases, the same dendrites freely interact with the dendrites of neighboring neurons of the same type; this is called ‘self/non-self discrimination’. It is not clear, however, how these two processes influence the activity of neural circuits. Both self-avoidance and self/non-self discrimination rely on the expression of genes that encode so-called recognition molecules. Kostadinov and Sanes have now altered the expression of these genes in mice to see the effect that disrupting these two phenomena has on a set of neurons called ‘starburst amacrine cells’ that are found at the back the eye. The dendrites of starburst amacrine cells generate signals when objects move across the animal's field of vision. These dendrites then signal to other starburst amacrine cells and to so-called ‘direction-selective ganglion cells’, which in turn send this information to the brain for further processing. The experiments revealed that these disruptions affected the connections between the dendrites. Starburst amacrine cells that lacked self-avoidance mistakenly formed connections with themselves—as if they mistook their own dendrites for those of other starburst cells. In contrast, neurons that lacked self/non-self discrimination made the opposite mistake, and rarely formed connections with each other—as if they mistook the dendrites of other starbursts for their own. Disruptions to either phenomenon interfered with the activity of the direction-selective ganglion cells. Following on from the work of Kostadinov and Sanes, the next challenges include uncovering how the recognition molecules help with self-avoidance and self/non-self discrimination. It will also be important to examine whether the conclusions based on one type of neurons can be generalized to others that also exhibit these two phenomena. DOI:http://dx.doi.org/10.7554/eLife.08964.002
Collapse
Affiliation(s)
- Dimitar Kostadinov
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Joshua R Sanes
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| |
Collapse
|
25
|
Netrin-4 regulates thalamocortical axon branching in an activity-dependent fashion. Proc Natl Acad Sci U S A 2014; 111:15226-31. [PMID: 25288737 DOI: 10.1073/pnas.1402095111] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Axon branching is remodeled by sensory-evoked and spontaneous neuronal activity. However, the underlying molecular mechanism is largely unknown. Here, we demonstrate that the netrin family member netrin-4 (NTN4) contributes to activity-dependent thalamocortical (TC) axon branching. In the postnatal developmental stages of rodents, ntn4 expression was abundant in and around the TC recipient layers of sensory cortices. Neuronal activity dramatically altered the ntn4 expression level in the cortex in vitro and in vivo. TC axon branching was promoted by exogenous NTN4 and suppressed by depletion of the endogenous protein. Moreover, unc-5 homolog B (Unc5B), which strongly bound to NTN4, was expressed in the sensory thalamus, and knockdown of Unc5B in thalamic cells markedly reduced TC axon branching. These results suggest that NTN4 acts as a positive regulator for TC axon branching through activity-dependent expression.
Collapse
|
26
|
Calsyntenin-1 regulates axon branching and endosomal trafficking during sensory neuron development in vivo. J Neurosci 2014; 34:9235-48. [PMID: 25009257 DOI: 10.1523/jneurosci.0561-14.2014] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Precise regulation of axon branching is crucial for neuronal circuit formation, yet the mechanisms that control branch formation are not well understood. Moreover, the highly complex morphology of neurons makes them critically dependent on protein/membrane trafficking and transport systems, although the functions for membrane trafficking in neuronal morphogenesis are largely undefined. Here we identify a kinesin adaptor, Calsyntenin-1 (Clstn-1), as an essential regulator of axon branching and neuronal compartmentalization in vivo. We use morpholino knockdown and a Clstn-1 mutant to show that Clstn-1 is required for formation of peripheral but not central sensory axons, and for peripheral axon branching in zebrafish. We used live imaging of endosomal trafficking in vivo to show that Clstn-1 regulates transport of Rab5-containing endosomes from the cell body to specific locations of developing axons. Our results suggest a model in which Clstn-1 patterns separate axonal compartments and define their ability to branch by directing trafficking of specific endosomes.
Collapse
|
27
|
Moreno RL, Ribera AB. Spinal neurons require Islet1 for subtype-specific differentiation of electrical excitability. Neural Dev 2014; 9:19. [PMID: 25149090 PMCID: PMC4153448 DOI: 10.1186/1749-8104-9-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 07/16/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the spinal cord, stereotypic patterns of transcription factor expression uniquely identify neuronal subtypes. These transcription factors function combinatorially to regulate gene expression. Consequently, a single transcription factor may regulate divergent development programs by participation in different combinatorial codes. One such factor, the LIM-homeodomain transcription factor Islet1, is expressed in the vertebrate spinal cord. In mouse, chick and zebrafish, motor and sensory neurons require Islet1 for specification of biochemical and morphological signatures. Little is known, however, about the role that Islet1 might play for development of electrical membrane properties in vertebrates. Here we test for a role of Islet1 in differentiation of excitable membrane properties of zebrafish spinal neurons. RESULTS We focus our studies on the role of Islet1 in two populations of early born zebrafish spinal neurons: ventral caudal primary motor neurons (CaPs) and dorsal sensory Rohon-Beard cells (RBs). We take advantage of transgenic lines that express green fluorescent protein (GFP) to identify CaPs, RBs and several classes of interneurons for electrophysiological study. Upon knock-down of Islet1, cells occupying CaP-like and RB-like positions continue to express GFP. With respect to voltage-dependent currents, CaP-like and RB-like neurons have novel repertoires that distinguish them from control CaPs and RBs, and, in some respects, resemble those of neighboring interneurons. The action potentials fired by CaP-like and RB-like neurons also have significantly different properties compared to those elicited from control CaPs and RBs. CONCLUSIONS Overall, our findings suggest that, for both ventral motor and dorsal sensory neurons, Islet1 directs differentiation programs that ultimately specify electrical membrane as well as morphological properties that act together to sculpt neuron identity.
Collapse
Affiliation(s)
- Rosa L Moreno
- Department of Physiology, University of Colorado Anschutz Medical Campus, RC-1 North, 7403A, Mailstop 8307, 12800 E 19th Ave,, 80045 Aurora, CO, USA.
| | | |
Collapse
|
28
|
Gibson DA, Tymanskyj S, Yuan RC, Leung HC, Lefebvre JL, Sanes JR, Chédotal A, Ma L. Dendrite self-avoidance requires cell-autonomous slit/robo signaling in cerebellar purkinje cells. Neuron 2014; 81:1040-1056. [PMID: 24607227 DOI: 10.1016/j.neuron.2014.01.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
Dendrites from the same neuron usually develop nonoverlapping patterns by self-avoidance, a process requiring contact-dependent recognition and repulsion. Recent studies have implicated homophilic interactions of cell surface molecules, including Dscams and Pcdhgs, in self-recognition, but repulsive molecular mechanisms remain obscure. Here, we report a role for the secreted molecule Slit2 and its receptor Robo2 in self-avoidance of cerebellar Purkinje cells (PCs). Both molecules are highly expressed by PCs, and their deletion leads to excessive dendrite self-crossing without affecting arbor size and shape. This cell-autonomous function is supported by the boundary-establishing activity of Slit in culture and the phenotype rescue by membrane-associated Slit2 activities. Furthermore, genetic studies show that they act independently from Pcdhg-mediated recognition. Finally, PC-specific deletion of Robo2 is associated with motor behavior alterations. Thus, our study uncovers a local repulsive mechanism required for self-avoidance and demonstrates the molecular complexity at the cell surface in dendritic patterning.
Collapse
Affiliation(s)
- Daniel A Gibson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Stephen Tymanskyj
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Rachel C Yuan
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Haiwen C Leung
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Julie L Lefebvre
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alain Chédotal
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMR_S968, CNRS_UMR7210, Institut de la Vision, 750012, Paris, France
| | - Le Ma
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
29
|
Orlic-Milacic M, Kaufman L, Mikhailov A, Cheung AYL, Mahmood H, Ellis J, Gianakopoulos PJ, Minassian BA, Vincent JB. Over-expression of either MECP2_e1 or MECP2_e2 in neuronally differentiated cells results in different patterns of gene expression. PLoS One 2014; 9:e91742. [PMID: 24699272 PMCID: PMC3974668 DOI: 10.1371/journal.pone.0091742] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 02/14/2014] [Indexed: 02/01/2023] Open
Abstract
Mutations in MECP2 are responsible for the majority of Rett syndrome cases. MECP2 is a regulator of transcription, and has two isoforms, MECP2_e1 and MECP2_e2. There is accumulating evidence that MECP2_e1 is the etiologically relevant variant for Rett. In this study we aim to detect genes that are differentially transcribed in neuronal cells over-expressing either of these two MECP2 isoforms. The human neuroblastoma cell line SK-N-SH was stably infected by lentiviral vectors over-expressing MECP2_e1, MECP2_e2, or eGFP, and were then differentiated into neurons. The same lentiviral constructs were also used to infect mouse Mecp2 knockout (Mecp2tm1.1Bird) fibroblasts. RNA from these cells was used for microarray gene expression analysis. For the human neuronal cells, ∼800 genes showed >three-fold change in expression level with the MECP2_e1 construct, and ∼230 with MECP2_e2 (unpaired t-test, uncorrected p value <0.05). We used quantitative RT-PCR to verify microarray results for 41 of these genes. We found significant up-regulation of several genes resulting from over-expression of MECP2_e1 including SRPX2, NAV3, NPY1R, SYN3, and SEMA3D. DOCK8 was shown via microarray and qRT-PCR to be upregulated in both SK-N-SH cells and mouse fibroblasts. Both isoforms up-regulated GABRA2, KCNA1, FOXG1 and FOXP2. Down-regulation of expression in the presence of MECP2_e1 was seen with UNC5C and RPH3A. Understanding the biology of these differentially transcribed genes and their role in neurodevelopment may help us to understand the relative functions of the two MECP2 isoforms, and ultimately develop a better understanding of RTT etiology and determine the clinical relevance of isoform-specific mutations.
Collapse
Affiliation(s)
- Marija Orlic-Milacic
- Molecular Neuropsychiatry & Development Lab, Campbell Family Mental Health Research Institute, The Centre for Addiction & Mental Health, Toronto, Ontario, Canada
| | - Liana Kaufman
- Molecular Neuropsychiatry & Development Lab, Campbell Family Mental Health Research Institute, The Centre for Addiction & Mental Health, Toronto, Ontario, Canada
| | - Anna Mikhailov
- Molecular Neuropsychiatry & Development Lab, Campbell Family Mental Health Research Institute, The Centre for Addiction & Mental Health, Toronto, Ontario, Canada
| | - Aaron Y. L. Cheung
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Huda Mahmood
- Molecular Neuropsychiatry & Development Lab, Campbell Family Mental Health Research Institute, The Centre for Addiction & Mental Health, Toronto, Ontario, Canada
| | - James Ellis
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Peter J. Gianakopoulos
- Molecular Neuropsychiatry & Development Lab, Campbell Family Mental Health Research Institute, The Centre for Addiction & Mental Health, Toronto, Ontario, Canada
| | - Berge A. Minassian
- Program in Genetics & Genomic Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - John B. Vincent
- Molecular Neuropsychiatry & Development Lab, Campbell Family Mental Health Research Institute, The Centre for Addiction & Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
30
|
Branch management: mechanisms of axon branching in the developing vertebrate CNS. Nat Rev Neurosci 2014; 15:7-18. [PMID: 24356070 DOI: 10.1038/nrn3650] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The remarkable ability of a single axon to extend multiple branches and form terminal arbors enables vertebrate neurons to integrate information from divergent regions of the nervous system. Axons select appropriate pathways during development, but it is the branches that extend interstitially from the axon shaft and arborize at specific targets that are responsible for virtually all of the synaptic connectivity in the vertebrate CNS. How do axons form branches at specific target regions? Recent studies have identified molecular cues that activate intracellular signalling pathways in axons and mediate dynamic reorganization of the cytoskeleton to promote the formation of axon branches.
Collapse
|
31
|
New insights into the roles of the contactin cell adhesion molecules in neural development. ADVANCES IN NEUROBIOLOGY 2014; 8:165-94. [PMID: 25300137 DOI: 10.1007/978-1-4614-8090-7_8] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In vertebrates, the contactin (CNTN) family of neural cell recognition molecules includes six related cell adhesion molecules that play non-overlapping roles in the formation and maintenance of the nervous system. CNTN1 and CNTN2 are the prototypical members of the family and have been involved, through cis- and trans-interactions with distinct cell adhesion molecules, in neural cell migration, axon guidance, and the organization of myelin subdomains. In contrast, the roles of CNTN3-6 are less well characterized although the generation of null mice and the recent identification of a common extracellular binding partner have considerably advanced our grasp of their physiological roles in particular as they relate to the wiring of sensory tissues. In this review, we aim to present a summary of our current understanding of CNTN functions and give an overview of the challenges that lie ahead in understanding the roles these proteins play in nervous system development and maintenance.
Collapse
|
32
|
Wang F, Julien DP, Sagasti A. Journey to the skin: Somatosensory peripheral axon guidance and morphogenesis. Cell Adh Migr 2013; 7:388-94. [PMID: 23670092 PMCID: PMC3739816 DOI: 10.4161/cam.25000] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The peripheral axons of vertebrate tactile somatosensory neurons travel long distances from ganglia just outside the central nervous system to the skin. Once in the skin these axons form elaborate terminals whose organization must be regionally patterned to detect and accurately localize different kinds of touch stimuli. This review describes key studies that identified choice points for somatosensory axon growth cones and the extrinsic molecular cues that function at each of those steps. While much has been learned in the past 20 years about the guidance of these axons, there is still much to be learned about how the peripheral axons of different kinds of somatosensory neurons adopt different trajectories and form specific terminal structures.
Collapse
Affiliation(s)
- Fang Wang
- Department of Molecular, Cell and Developmental Biology; University of California, Los Angeles; Los Angeles, CA USA
| | | | | |
Collapse
|
33
|
Chen Z, Lee H, Henle SJ, Cheever TR, Ekker SC, Henley JR. Primary neuron culture for nerve growth and axon guidance studies in zebrafish (Danio rerio). PLoS One 2013; 8:e57539. [PMID: 23469201 PMCID: PMC3587632 DOI: 10.1371/journal.pone.0057539] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/25/2013] [Indexed: 12/21/2022] Open
Abstract
Zebrafish (Danio rerio) is a widely used model organism in genetics and developmental biology research. Genetic screens have proven useful for studying embryonic development of the nervous system in vivo, but in vitro studies utilizing zebrafish have been limited. Here, we introduce a robust zebrafish primary neuron culture system for functional nerve growth and guidance assays. Distinct classes of central nervous system neurons from the spinal cord, hindbrain, forebrain, and retina from wild type zebrafish, and fluorescent motor neurons from transgenic reporter zebrafish lines, were dissociated and plated onto various biological and synthetic substrates to optimize conditions for axon outgrowth. Time-lapse microscopy revealed dynamically moving growth cones at the tips of extending axons. The mean rate of axon extension in vitro was 21.4±1.2 µm hr−1 s.e.m. for spinal cord neurons, which corresponds to the typical ∼0.5 mm day−1 growth rate of nerves in vivo. Fluorescence labeling and confocal microscopy demonstrated that bundled microtubules project along axons to the growth cone central domain, with filamentous actin enriched in the growth cone peripheral domain. Importantly, the growth cone surface membrane expresses receptors for chemotropic factors, as detected by immunofluorescence microscopy. Live-cell functional assays of axon extension and directional guidance demonstrated mammalian brain-derived neurotrophic factor (BDNF)-dependent stimulation of outgrowth and growth cone chemoattraction, whereas mammalian myelin-associated glycoprotein inhibited outgrowth. High-resolution live-cell Ca2+-imaging revealed local elevation of cytoplasmic Ca2+ concentration in the growth cone induced by BDNF application. Moreover, BDNF-induced axon outgrowth, but not basal outgrowth, was blocked by treatments to suppress cytoplasmic Ca2+ signals. Thus, this primary neuron culture model system may be useful for studies of neuronal development, chemotropic axon guidance, and mechanisms underlying inhibition of neural regeneration in vitro, and complement observations made in vivo.
Collapse
Affiliation(s)
- Zheyan Chen
- Mayo Graduate School, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Han Lee
- Mayo Graduate School, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Steven J. Henle
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Thomas R. Cheever
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Stephen C. Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - John R. Henley
- Mayo Graduate School, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
34
|
Baudet ML, Bellon A, Holt CE. Role of microRNAs in Semaphorin function and neural circuit formation. Semin Cell Dev Biol 2012; 24:146-55. [PMID: 23219835 DOI: 10.1016/j.semcdb.2012.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 10/19/2012] [Accepted: 11/28/2012] [Indexed: 01/23/2023]
Abstract
Since the discovery of the first microRNA (miRNA) almost 20 years ago, insight into their functional role has gradually been accumulating. This class of non-coding RNAs has recently been implicated as key molecular regulators in the biology of most eukaryotic cells, contributing to the physiology of various systems including immune, cardiovascular, nervous systems and also to the pathophysiology of cancers. Interestingly, Semaphorins, a class of evolutionarily conserved signalling molecules, are acknowledged to play major roles in these systems also. This, combined with the fact that Semaphorin signalling requires tight spatiotemporal regulation, a hallmark of miRNA expression, suggests that miRNAs could be crucial regulators of Semaphorin function. Here, we review evidence suggesting that Semaphorin signalling is regulated by miRNAs in various systems in health and disease. In particular, we focus on neural circuit formation, including axon guidance, where Semaphorin function was first discovered.
Collapse
|
35
|
Santiago-Medina M, Myers JP, Gomez TM. Imaging adhesion and signaling dynamics in Xenopus laevis growth cones. Dev Neurobiol 2012; 72:585-99. [PMID: 21465668 DOI: 10.1002/dneu.20886] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Xenopus laevis provides a robust model system to study cellular signaling and downstream processes during development both in vitro and in vivo. Intracellular signals must function within highly restricted spatial and temporal domains to activate specific downstream targets and cellular processes. Combining the versatility of developing Xenopus neurons with advances in fluorescent protein biosensors and imaging technologies has allowed many dynamic cellular processes to be visualized. This review will focus on the techniques we use to visualize and measure cell signaling, motility and adhesion by quantitative fluorescence microscopy in vitro and in vivo.
Collapse
Affiliation(s)
- Miguel Santiago-Medina
- Department of Neuroscience, Neuroscience Training Program, University of Wisconsin-Madison, WI 53706, USA
| | | | | |
Collapse
|
36
|
Roberts A, Li WC, Soffe SR. A functional scaffold of CNS neurons for the vertebrates: the developing Xenopus laevis spinal cord. Dev Neurobiol 2012; 72:575-84. [PMID: 21485014 DOI: 10.1002/dneu.20889] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In young and developing amphibians and fish the spinal cord is functional but remarkably simple compared with the adult. Is the pattern of neurons and their connections common across at least these lower vertebrates? Does this basic pattern extend into the brainstem? Could the development of simple functioning neuronal networks depend on very basic rules of connectivity and act as pioneer networks providing a substrate for the development of more complex and subtle networks. In this review of the functional neuron classes in the Xenopus laevis tadpole spinal cord up to hatching, we will consider progress and difficulties in using anatomy, transcription factor expression, physiology, and activity to define spinal neuron types. Even here it is not straightforward and is rarely possible to bring all the different strands of evidence together. But, we think we have a rather complete picture of the hatchling tadpole spinal neuron types and can define clear roles for most of them in behavior. Our present knowledge about the hatchling Xenopus spinal cord should set up many of the problems to be unraveled in the future by more developmentally oriented research.
Collapse
Affiliation(s)
- Alan Roberts
- Biological Sciences, University of Bristol, Woodland Road, Bristol, United Kingdom.
| | | | | |
Collapse
|
37
|
Hung FC, Cheng YC, Sun NK, Chao CCK. Identification and functional characterization of zebrafish Gas7 gene in early development. J Neurosci Res 2012; 91:51-61. [PMID: 23086717 DOI: 10.1002/jnr.23145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 08/07/2012] [Accepted: 08/22/2012] [Indexed: 12/17/2022]
Abstract
Growth arrest-specific 7 (Gas7) is preferentially expressed in the nervous system and plays an important role during neuritogenesis in mammals. However, the structure and function of Gas7 homologs have not been studied in nonmammalian vertebrates used as models. In this report, we identify a Gas7 gene in zebrafish that we termed zfGas7. The transcript of this gene was produced by canonical splicing, and its protein product contained a Fes/CIP4 homology and a coiled-coil domain. In early zebrafish embryos, RT-PCR analyses revealed that zfGas7 was initially expressed at 5.3 hr postfertilization (hpf), followed by an increase of expression at 10 hpf and further accumulation during somitogenesis at 48 hpf. Spatiotemporal analyses further showed that Gas7 mRNA was detected in the brain, somite, and posterior presomitic mesoderm regions during somitogenesis. At 36 hpf, zfGas7 mRNA was detected in the brain and somite but was later found only in neuronal clusters of the brain at 52 hpf. Gas7 knockdown with morpholino antisense oligonucleotides (Gas7MO) reduced the number of HuC-positive neurons in the trigeminal and statoacoustic ganglions and produced deformed phenotypes, such as flattening of the top of the head. Notably, the neuron reduction and deformed phenotypes observed in Gas7MO embryos were partially rescued by ectopic expression of Gas7. Because altered somitogenesis and pigmentation were also found in the morphants, the neuronal phenotypes observed likely are due to a general developmental delay of embryogenesis. These results indicate that Gas7 is expressed in neuronal cells but is not specifically required for neuronal development in vertebrates.
Collapse
Affiliation(s)
- Feng-Chun Hung
- Department of Biochemistry and Molecular Biology, Chang Gung University, Gueishan, Taiwan, Republic of China
| | | | | | | |
Collapse
|
38
|
Tanaka H, Morimura R, Ohshima T. Dpysl2 (CRMP2) and Dpysl3 (CRMP4) phosphorylation by Cdk5 and DYRK2 is required for proper positioning of Rohon-Beard neurons and neural crest cells during neurulation in zebrafish. Dev Biol 2012; 370:223-36. [PMID: 22898304 DOI: 10.1016/j.ydbio.2012.07.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 07/21/2012] [Accepted: 07/31/2012] [Indexed: 11/27/2022]
Abstract
Dpysl2 (CRMP2) and Dpysl3 (CRMP4) are involved in neuronal polarity and axon elongation in cultured neurons. These proteins are expressed in various regions of the developing nervous system, but their roles in vivo are largely unknown. In dpysl2 and dpysl3 double morphants, Rohon-Beard (RB) primary sensory neurons that were originally located bilaterally along the midline shifted their position to a more medial location in the dorsal-most part of spinal cord. A similar phenotype was observed in the cdk5 and dyrk2 double morphants. Dpysl2 and Dpysl3 phosphorylation mimics recovered this phenotype. Cell transplantation analysis demonstrated that this ectopic RB cell positioning was non-cell autonomous and correlated with the abnormal position of neural crest cells (NCCs), which also occupied the dorsal-most part of the spinal cord during the neural rod formation stage. The cell position of other interneuron and motor neurons within the central nervous system was normal in these morphants. These results suggest that the phosphorylation of Dpysl2 and Dpysl3 by Cdk5 and DYRK2 is required for the proper positioning of RB neurons and NCCs during neurulation in zebrafish embryos.
Collapse
Affiliation(s)
- Hideomi Tanaka
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162 8480, Japan
| | | | | |
Collapse
|
39
|
Wang F, Wolfson SN, Gharib A, Sagasti A. LAR receptor tyrosine phosphatases and HSPGs guide peripheral sensory axons to the skin. Curr Biol 2012; 22:373-82. [PMID: 22326027 PMCID: PMC3298620 DOI: 10.1016/j.cub.2012.01.040] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 01/06/2012] [Accepted: 01/20/2012] [Indexed: 11/20/2022]
Abstract
BACKGROUND Peripheral axons of somatosensory neurons innervate the skin early in development to detect touch stimuli. Embryological experiments had suggested that the skin produces guidance cues that attract sensory axons, but neither the attractants nor their neuronal receptors had previously been identified. RESULTS To investigate peripheral axon navigation to the skin, we combined live imaging of developing zebrafish Rohon-Beard (RB) neurons with molecular loss-of-function manipulations. Simultaneously knocking down two members of the leukocyte antigen-related (LAR) family of receptor tyrosine phosphatases expressed in RB neurons, or inhibiting their function with dominant-negative proteins, misrouted peripheral axons to internal tissues. Time-lapse imaging indicated that peripheral axon guidance, rather than outgrowth or maintenance, was defective in LAR-deficient neurons. Peripheral axons displayed a similar misrouting phenotype in mutants defective in heparan sulfate proteoglycan (HSPG) production and avoided regions in which HSPGs were locally degraded. CONCLUSIONS HSPGs and LAR family receptors are required for sensory axon guidance to the skin. Together, our results support a model in which peripheral HSPGs are attractive ligands for LAR receptors on RB neurons.
Collapse
Affiliation(s)
- Fang Wang
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | | | | | | |
Collapse
|
40
|
Abstract
Axon branching is a complex morphological process, the regulation of which we are just beginning to understand. Many factors known to be important for axon growth and guidance have emerged as key regulators of axon branching. The extrinsic factors implicated in axon branching include traditional axon guidance cues such as the slits, semaphorins, and ephrins; neurotrophins such as BDNF; the secreted glycoprotein Wnt; the extracellular matrix protein anosmin-1; and certain transmembrane cell adhesion molecules--as well as sensory experience and neuronal activity. Although less is known about the intracellular control of axon branching, in recent years significant advances have been made in this area. Kinases and their regulators, Rho GTPases and their regulators, transcription factors, ubiquitin ligases, and several microtubule and actin-binding proteins are now implicated in the control of axon branching. It is likely that many more branching regulators remain to be discovered, as do the links between extrinsic cues and intracellular signaling proteins in the control of axon branching.
Collapse
Affiliation(s)
- Parizad M Bilimoria
- Department of Neurobiology and Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
41
|
Stockinger P, Maître JL, Heisenberg CP. Defective neuroepithelial cell cohesion affects tangential branchiomotor neuron migration in the zebrafish neural tube. Development 2011; 138:4673-83. [PMID: 21965614 DOI: 10.1242/dev.071233] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Facial branchiomotor neurons (FBMNs) in zebrafish and mouse embryonic hindbrain undergo a characteristic tangential migration from rhombomere (r) 4, where they are born, to r6/7. Cohesion among neuroepithelial cells (NCs) has been suggested to function in FBMN migration by inhibiting FBMNs positioned in the basal neuroepithelium such that they move apically between NCs towards the midline of the neuroepithelium instead of tangentially along the basal side of the neuroepithelium towards r6/7. However, direct experimental evaluation of this hypothesis is still lacking. Here, we have used a combination of biophysical cell adhesion measurements and high-resolution time-lapse microscopy to determine the role of NC cohesion in FBMN migration. We show that reducing NC cohesion by interfering with Cadherin 2 (Cdh2) activity results in FBMNs positioned at the basal side of the neuroepithelium moving apically towards the neural tube midline instead of tangentially towards r6/7. In embryos with strongly reduced NC cohesion, ectopic apical FBMN movement frequently results in fusion of the bilateral FBMN clusters over the apical midline of the neural tube. By contrast, reducing cohesion among FBMNs by interfering with Contactin 2 (Cntn2) expression in these cells has little effect on apical FBMN movement, but reduces the fusion of the bilateral FBMN clusters in embryos with strongly diminished NC cohesion. These data provide direct experimental evidence that NC cohesion functions in tangential FBMN migration by restricting their apical movement.
Collapse
Affiliation(s)
- Petra Stockinger
- Institute of Science and Technology Austria, Am Campus 1, A-3400 Klosterneuburg, Austria
| | | | | |
Collapse
|
42
|
Hung RJ, Terman JR. Extracellular inhibitors, repellents, and semaphorin/plexin/MICAL-mediated actin filament disassembly. Cytoskeleton (Hoboken) 2011; 68:415-33. [PMID: 21800438 PMCID: PMC3612987 DOI: 10.1002/cm.20527] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 07/21/2011] [Indexed: 01/29/2023]
Abstract
Multiple extracellular signals have been identified that regulate actin dynamics within motile cells, but how these instructive cues present on the cell surface exert their precise effects on the internal actin cytoskeleton is still poorly understood. One particularly interesting class of these cues is a group of extracellular proteins that negatively alter the movement of cells and their processes. Over the years, these types of events have been described using a variety of terms and herein we provide an overview of inhibitory/repulsive cellular phenomena and highlight the largest known protein family of repulsive extracellular cues, the Semaphorins. Specifically, the Semaphorins (Semas) utilize Plexin cell-surface receptors to dramatically collapse the actin cytoskeleton and we summarize what is known of the direct molecular and biochemical mechanisms of Sema-triggered actin filament (F-actin) disassembly. We also discuss new observations from our lab that reveal that the multidomain oxidoreductase (Redox) enzyme Molecule Interacting with CasL (MICAL), an important mediator of Sema/Plexin repulsion, is a novel F-actin disassembly factor. Our results indicate that MICAL triggers Sema/Plexin-mediated reorganization of the F-actin cytoskeleton and suggest a role for specific Redox signaling events in regulating actin dynamics.
Collapse
Affiliation(s)
- Ruei-Jiun Hung
- Departments of Neuroscience and Pharmacology, and Neuroscience Graduate Program, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan R. Terman
- Departments of Neuroscience and Pharmacology, and Neuroscience Graduate Program, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
43
|
Andersen EF, Asuri NS, Halloran MC. In vivo imaging of cell behaviors and F-actin reveals LIM-HD transcription factor regulation of peripheral versus central sensory axon development. Neural Dev 2011; 6:27. [PMID: 21619654 PMCID: PMC3121664 DOI: 10.1186/1749-8104-6-27] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 05/27/2011] [Indexed: 02/01/2023] Open
Abstract
Background Development of specific neuronal morphology requires precise control over cell motility processes, including axon formation, outgrowth and branching. Dynamic remodeling of the filamentous actin (F-actin) cytoskeleton is critical for these processes; however, little is known about the mechanisms controlling motile axon behaviors and F-actin dynamics in vivo. Neuronal structure is specified in part by intrinsic transcription factor activity, yet the molecular and cellular steps between transcription and axon behavior are not well understood. Zebrafish Rohon-Beard (RB) sensory neurons have a unique morphology, with central axons that extend in the spinal cord and a peripheral axon that innervates the skin. LIM homeodomain (LIM-HD) transcription factor activity is required for formation of peripheral RB axons. To understand how neuronal morphogenesis is controlled in vivo and how LIM-HD transcription factor activity differentially regulates peripheral versus central axons, we used live imaging of axon behavior and F-actin distribution in vivo. Results We used an F-actin biosensor containing the actin-binding domain of utrophin to characterize actin rearrangements during specific developmental processes in vivo, including axon initiation, consolidation and branching. We found that peripheral axons initiate from a specific cellular compartment and that F-actin accumulation and protrusive activity precede peripheral axon initiation. Moreover, disruption of LIM-HD transcriptional activity has different effects on the motility of peripheral versus central axons; it inhibits peripheral axon initiation, growth and branching, while increasing the growth rate of central axons. Our imaging revealed that LIM-HD transcription factor activity is not required for F-actin based protrusive activity or F-actin accumulation during peripheral axon initiation, but can affect positioning of F-actin accumulation and axon formation. Conclusion Our ability to image the dynamics of F-actin distribution during neuronal morphogenesis in vivo is unprecedented, and our experiments provide insight into the regulation of cell motility as neurons develop in the intact embryo. We identify specific motile cell behaviors affected by LIM-HD transcription factor activity and reveal how transcription factors differentially control the formation and growth of two axons from the same neuron.
Collapse
Affiliation(s)
- Erica F Andersen
- Genetics Training Program, University of Wisconsin, 1117 W, Johnson Street, Madison, WI 53706, USA
| | | | | |
Collapse
|
44
|
Rieger S, Wang F, Sagasti A. Time-lapse imaging of neural development: zebrafish lead the way into the fourth dimension. Genesis 2011; 49:534-45. [PMID: 21305690 DOI: 10.1002/dvg.20729] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/24/2011] [Accepted: 01/25/2011] [Indexed: 01/01/2023]
Abstract
Time-lapse imaging is often the only way to appreciate fully the many dynamic cell movements critical to neural development. Zebrafish possess many advantages that make them the best vertebrate model organism for live imaging of dynamic development events. This review will discuss technical considerations of time-lapse imaging experiments in zebrafish, describe selected examples of imaging studies in zebrafish that revealed new features or principles of neural development, and consider the promise and challenges of future time-lapse studies of neural development in zebrafish embryos and adults.
Collapse
Affiliation(s)
- Sandra Rieger
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California, USA
| | | | | |
Collapse
|
45
|
Gibson DA, Ma L. Developmental regulation of axon branching in the vertebrate nervous system. Development 2011; 138:183-95. [PMID: 21177340 DOI: 10.1242/dev.046441] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During nervous system development, axons generate branches to connect with multiple synaptic targets. As with axon growth and guidance, axon branching is tightly controlled in order to establish functional neural circuits, yet the mechanisms that regulate this important process are less well understood. Here, we review recent advances in the study of several common branching processes in the vertebrate nervous system. By focusing on each step in these processes we illustrate how different types of branching are regulated by extracellular cues and neural activity, and highlight some common principles that underlie the establishment of complex neural circuits in vertebrate development.
Collapse
Affiliation(s)
- Daniel A Gibson
- Zilkha Neurogenetic Institute, Department of Cell and Neurobiology, Keck School of Medicine, Neuroscience Graduate Program, University of Southern California, 1501 San Pablo Street, Los Angeles, CA 90089, USA
| | | |
Collapse
|
46
|
Fujii T, Uchiyama H, Yamamoto N, Hori H, Tatsumi M, Ishikawa M, Arima K, Higuchi T, Kunugi H. Possible association of the semaphorin 3D gene (SEMA3D) with schizophrenia. J Psychiatr Res 2011; 45:47-53. [PMID: 20684831 DOI: 10.1016/j.jpsychires.2010.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 04/01/2010] [Accepted: 05/06/2010] [Indexed: 10/19/2022]
Abstract
Semaphorins are ligands of plexins, and the plexin-semaphorin signaling system is widely involved in many neuronal events including axon guidance, cell migration, axon pruning, and synaptic plasticity. The plexin A2 gene (PLXNA2) has been reported to be associated with schizophrenia. This finding prompted us to examine the possible association between the semaphorin 3D gene (SEMA3D) and schizophrenia in a Japanese population. We genotyped 9 tagging single nucleotide polymorphisms (SNPs) of SEMA3D including a non-synonymous variation, Lys701Gln (rs7800072), in a sample of 506 patients with schizophrenia and 941 healthy control subjects. The Gln701 allele showed a significant protective effect against the development of schizophrenia (p = 0.0069, odds ratio = 0.76, 95% confidence interval 0.63 to 0.93). Furthermore, the haplotype-based analyses revealed a significant association. The four-marker analysis (rs2190208-rs1029564-rs17159614-rs12176601), in particular, not including the Lys701Gln, revealed a highly significant association (p = 0.00001, global permutation), suggesting that there may be other functional polymorphisms within SEMA3D. Our findings provide strong evidence that SEMA3D confers susceptibility to schizophrenia, which could contribute to the neurodevelopmental impairments in the disorder.
Collapse
Affiliation(s)
- Takashi Fujii
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Tanaka H, Nojima Y, Shoji W, Sato M, Nakayama R, Ohshima T, Okamoto H. Islet1 selectively promotes peripheral axon outgrowth in Rohon-Beard primary sensory neurons. Dev Dyn 2010; 240:9-22. [DOI: 10.1002/dvdy.22499] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
48
|
The expression of TAG-1 in glial cells is sufficient for the formation of the juxtaparanodal complex and the phenotypic rescue of tag-1 homozygous mutants in the CNS. J Neurosci 2010; 30:13943-54. [PMID: 20962216 DOI: 10.1523/jneurosci.2574-10.2010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Myelinated fibers are organized into specialized domains that ensure the rapid propagation of action potentials and are characterized by protein complexes underlying axoglial interactions. TAG-1 (Transient Axonal Glycoprotein-1), a cell adhesion molecule of the Ig superfamily, is expressed by neurons as well as by myelinating glia. It is essential for the molecular organization of myelinated fibers as it maintains the integrity of the juxtaparanodal region through its interactions with Caspr2 and the voltage-gated potassium channels (VGKCs) on the axolemma. Since TAG-1 is the only known component of the juxtaparanodal complex expressed by the glial cell, it is important to clarify its role in the molecular organization of juxtaparanodes. For this purpose, we generated transgenic mice that exclusively express TAG-1 in oligodendrocytes and lack endogenous gene expression (Tag-1(-/-);plp(Tg(rTag-1))). Phenotypic analysis clearly demonstrates that glial TAG-1 is sufficient for the proper organization and maintenance of the juxtaparanodal domain in the CNS. Biochemical analysis shows that glial TAG-1 physically interacts with Caspr2 and VGKCs. Ultrastructural and behavioral analysis of Tag-1(-/-);plp(Tg(rTag-1)) mice shows that the expression of glial TAG-1 is sufficient to restore the axonal and myelin deficits as well as the behavioral defects observed in Tag-1(-/-) animals. Together, these data highlight the pivotal role of myelinating glia on axonal domain differentiation and organization.
Collapse
|
49
|
Martin SM, O'Brien GS, Portera-Cailliau C, Sagasti A. Wallerian degeneration of zebrafish trigeminal axons in the skin is required for regeneration and developmental pruning. Development 2010; 137:3985-94. [PMID: 21041367 DOI: 10.1242/dev.053611] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Fragments of injured axons that detach from their cell body break down by the molecularly regulated process of Wallerian degeneration (WD). Although WD resembles local axon degeneration, a common mechanism for refining neuronal structure, several previously examined instances of developmental pruning were unaffected by WD pathways. We used laser axotomy and time-lapse confocal imaging to characterize and compare peripheral sensory axon WD and developmental pruning in live zebrafish larvae. Detached fragments of single injured axon arbors underwent three stereotyped phases of WD: a lag phase, a fragmentation phase and clearance. The lag phase was developmentally regulated, becoming shorter as embryos aged, while the length of the clearance phase increased with the amount of axon debris. Both cell-specific inhibition of ubiquitylation and overexpression of the Wallerian degeneration slow protein (Wld(S)) lengthened the lag phase dramatically, but neither affected fragmentation. Persistent Wld(S)-expressing axon fragments directly repelled regenerating axon branches of their parent arbor, similar to self-repulsion among sister branches of intact arbors. Expression of Wld(S) also disrupted naturally occurring local axon pruning and axon degeneration in spontaneously dying trigeminal neurons: although pieces of Wld(S)-expressing axons were pruned, and some Wld(S)-expressing cells still died during development, in both cases detached axon fragments failed to degenerate. We propose that spontaneously pruned fragments of peripheral sensory axons must be removed by a WD-like mechanism to permit efficient innervation of the epidermis.
Collapse
Affiliation(s)
- Seanna M Martin
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
50
|
Cell Death as a Regulator of Cerebellar Histogenesis and Compartmentation. THE CEREBELLUM 2010; 10:373-92. [DOI: 10.1007/s12311-010-0222-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|