1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Nickerson KR, Sammoura FM, Zhou Y, Jaworski A. Slit-Robo signaling supports motor neuron avoidance of the spinal cord midline through DCC antagonism and other mechanisms. Front Cell Dev Biol 2025; 13:1563403. [PMID: 40276653 PMCID: PMC12018395 DOI: 10.3389/fcell.2025.1563403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Axon pathfinding and neuronal migration are orchestrated by attractive and repulsive guidance cues. In the mouse spinal cord, repulsion from Slit proteins through Robo family receptors and attraction to Netrin-1, mediated by the receptor DCC, control many aspects of neural circuit formation. This includes motor neuron wiring, where Robos help prevent both motor neuron cell bodies and axons from aberrantly crossing the spinal cord midline. These functions had been ascribed to Robo signaling being required to counter DCC-mediated attraction to Netrin-1 at the midline, either by mediating repulsion from midline-derived Slits or by silencing DCC signaling. However, the role of DCC in promoting motor neuron and axon midline crossing had not been directly tested. Here, we used in vivo mouse genetics and in vitro axon turning assays to further explore the interplay between Slit and Netrin signaling in motor neuron migration and axon guidance relative to the midline. We find that DCC is a major driver of midline crossing by motor axons, but not motor neuron cell bodies, when Robo1 and Robo2 are knocked out. Further, in vitro results indicate that Netrin-1 attracts motor axons and that Slits can modulate the chemotropic response to Netrin-1, converting it from attraction to repulsion. Our findings indicate that Robo signaling allows both motor neuron cell bodies and axons to avoid the midline, but that only motor axons require this pathway to antagonize DCC-dependent midline attraction, which likely involves a combination of mediating Slit repulsion and directly influencing Netrin-DCC signaling output.
Collapse
Affiliation(s)
- Kelsey R. Nickerson
- Department of Neuroscience, Brown University, Providence, RI, United States
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Ferass M. Sammoura
- Department of Neuroscience, Brown University, Providence, RI, United States
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Yonghong Zhou
- Department of Neuroscience, Brown University, Providence, RI, United States
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Alexander Jaworski
- Department of Neuroscience, Brown University, Providence, RI, United States
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, United States
| |
Collapse
|
3
|
Dailey-Krempel B, Martin AL, Jo HN, Junge HJ, Chen Z. A tug of war between DCC and ROBO1 signaling during commissural axon guidance. Cell Rep 2023; 42:112455. [PMID: 37149867 DOI: 10.1016/j.celrep.2023.112455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/07/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023] Open
Abstract
Dynamic and coordinated axonal responses to changing environments are critical for establishing neural connections. As commissural axons migrate across the CNS midline, they are suggested to switch from being attracted to being repelled in order to approach and to subsequently leave the midline. A molecular mechanism that is hypothesized to underlie this switch in axonal responses is the silencing of Netrin1/Deleted in Colorectal Carcinoma (DCC)-mediated attraction by the repulsive SLIT/ROBO1 signaling. Using in vivo approaches including CRISPR-Cas9-engineered mouse models of distinct Dcc splice isoforms, we show here that commissural axons maintain responsiveness to both Netrin and SLIT during midline crossing, although likely at quantitatively different levels. In addition, full-length DCC in collaboration with ROBO3 can antagonize ROBO1 repulsion in vivo. We propose that commissural axons integrate and balance the opposing DCC and Roundabout (ROBO) signaling to ensure proper guidance decisions during midline entry and exit.
Collapse
Affiliation(s)
| | - Andrew L Martin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ha-Neul Jo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Harald J Junge
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhe Chen
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
4
|
Godini R, Fallahi H, Pocock R. The regulatory landscape of neurite development in Caenorhabditis elegans. Front Mol Neurosci 2022; 15:974208. [PMID: 36090252 PMCID: PMC9453034 DOI: 10.3389/fnmol.2022.974208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/26/2022] [Indexed: 11/18/2022] Open
Abstract
Neuronal communication requires precise connectivity of neurite projections (axons and dendrites). Developing neurites express cell-surface receptors that interpret extracellular cues to enable correct guidance toward, and connection with, target cells. Spatiotemporal regulation of neurite guidance molecule expression by transcription factors (TFs) is critical for nervous system development and function. Here, we review how neurite development is regulated by TFs in the Caenorhabditis elegans nervous system. By collecting publicly available transcriptome and ChIP-sequencing data, we reveal gene expression dynamics during neurite development, providing insight into transcriptional mechanisms governing construction of the nervous system architecture.
Collapse
Affiliation(s)
- Rasoul Godini
- Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- *Correspondence: Rasoul Godini,
| | - Hossein Fallahi
- Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Roger Pocock
- Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Roger Pocock,
| |
Collapse
|
5
|
Gorla M, Bashaw GJ. Molecular mechanisms regulating axon responsiveness at the midline. Dev Biol 2020; 466:12-21. [PMID: 32818516 DOI: 10.1016/j.ydbio.2020.08.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
During embryonic development in bilaterally symmetric organisms, correct midline crossing is important for the proper formation of functional neural circuits. The aberrant development of neural circuits can result in multiple neurodevelopmental disorders, including horizontal gaze palsy, congenital mirror movement disorder, and autism spectrum disorder. Thus, understanding the molecular mechanisms that regulate proper axon guidance at the midline can provide insights into the pathology of neurological disorders. The signaling mechanisms that regulate midline crossing have been extensively studied in the Drosophila ventral nerve cord and the mouse embryonic spinal cord. In this review, we discuss these axon guidance mechanisms, highlighting the most recent advances in the understanding of how commissural axons switch their responsiveness from attractants to repellents during midline crossing.
Collapse
Affiliation(s)
- Madhavi Gorla
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Kaneyama T, Shirasaki R. Post-crossing segment of dI1 commissural axons forms collateral branches to motor neurons in the developing spinal cord. J Comp Neurol 2019; 526:1943-1961. [PMID: 29752714 DOI: 10.1002/cne.24464] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/30/2018] [Accepted: 05/03/2018] [Indexed: 11/09/2022]
Abstract
The dI1 commissural axons in the developing spinal cord, upon crossing the midline through the floor plate, make a sharp turn to grow rostrally. These post-crossing axons initially just extend adjacent to the floor plate without entering nearby motor columns. However, it remains poorly characterized how these post-crossing dI1 axons behave subsequently to this process. In the present study, to address this issue, we examined in detail the behavior of post-crossing dI1 axons in mice, using the Atoh1 enhancer-based conditional expression system that enables selective and sparse labeling of individual dI1 axons, together with Hb9 and ChAT immunohistochemistry for precise identification of spinal motor neurons (MNs). We found unexpectedly that the post-crossing segment of dI1 axons later gave off collateral branches that extended laterally to invade motor columns. Interestingly, these collateral branches emerged at around the time when their primary growth cones initiated invasion into motor columns. In addition, although the length of the laterally growing collateral branches increased with age, the majority of them remained within motor columns. Strikingly, these collateral branches further gave rise to multiple secondary branches in the region of MNs that innervate muscles close to the body axis. Moreover, these axonal branches formed presynaptic terminals on MNs. These observations demonstrate that dI1 commissural neurons develop axonal projection to spinal MNs via collateral branches arising later from the post-crossing segment of these axons. Our findings thus reveal a previously unrecognized projection of dI1 commissural axons that may contribute directly to generating proper motor output.
Collapse
Affiliation(s)
- Takeshi Kaneyama
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Ryuichi Shirasaki
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
7
|
Kinoshita-Kawada M, Hasegawa H, Hongu T, Yanagi S, Kanaho Y, Masai I, Mishima T, Chen X, Tsuboi Y, Rao Y, Yuasa-Kawada J, Wu JY. Explant Culture of the Embryonic Mouse Spinal Cord and Gene Transfer by ex vivo Electroporation. Bio Protoc 2019; 9:e3373. [PMID: 33654869 DOI: 10.21769/bioprotoc.3373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/15/2019] [Accepted: 08/18/2019] [Indexed: 11/02/2022] Open
Abstract
Developing axons change responsiveness to guidance cues during the journey to synapse with target cells. Axon crossing at the ventral midline serves as a model for studying how axons accomplish such a switch in their response. Although primary neuron culture has been a versatile technique for elucidating various developmental mechanisms, many in vivo characteristics of neurons, such as long axon-extending abilities and axonal compartments, are not thoroughly preserved. In explant cultures, such properties of differentiated neurons and tissue architecture are maintained. To examine how the midline repellent Slit regulated the distribution of the Robo receptor in spinal cord commissural axons upon midline crossing and whether Robo trafficking machinery was a determinant of midline crossing, novel explant culture systems were developed. We have combined an "open-book" spinal cord explant method with that devised for flat-mount retinae. Here we present our protocol for explant culture of embryonic mouse spinal cords, which allows flexible manipulation of experimental conditions, immunostaining of extending axons and quantitative analysis of individual axons. In addition, we present a modified method that combines ex vivo electroporation and "closed-book" spinal cord explant culture. These culture systems provide new platforms for detailed analysis of axon guidance, by adapting gene knockdown, knockout and genome editing.
Collapse
Affiliation(s)
- Mariko Kinoshita-Kawada
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan.,Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, Japan.,Department of Neurology, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hiroshi Hasegawa
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Hygienic Sciences, Kobe Pharmaceutical University, Kobe, Japan
| | - Tsunaki Hongu
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Heidelberg Institute for Stem Cell Technology and Experimental Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Yasunori Kanaho
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, Japan
| | - Takayasu Mishima
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Xiaoping Chen
- Department of Neurology, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yoshio Tsuboi
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yi Rao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University School of Life Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| | - Junichi Yuasa-Kawada
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan.,Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, Japan.,Department of Neurology, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Jane Y Wu
- Department of Neurology, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
8
|
Comer JD, Alvarez S, Butler SJ, Kaltschmidt JA. Commissural axon guidance in the developing spinal cord: from Cajal to the present day. Neural Dev 2019; 14:9. [PMID: 31514748 PMCID: PMC6739980 DOI: 10.1186/s13064-019-0133-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022] Open
Abstract
During neuronal development, the formation of neural circuits requires developing axons to traverse a diverse cellular and molecular environment to establish synaptic contacts with the appropriate postsynaptic partners. Essential to this process is the ability of developing axons to navigate guidance molecules presented by specialized populations of cells. These cells partition the distance traveled by growing axons into shorter intervals by serving as intermediate targets, orchestrating the arrival and departure of axons by providing attractive and repulsive guidance cues. The floor plate in the central nervous system (CNS) is a critical intermediate target during neuronal development, required for the extension of commissural axons across the ventral midline. In this review, we begin by giving a historical overview of the ventral commissure and the evolutionary purpose of decussation. We then review the axon guidance studies that have revealed a diverse assortment of midline guidance cues, as well as genetic and molecular regulatory mechanisms required for coordinating the commissural axon response to these cues. Finally, we examine the contribution of dysfunctional axon guidance to neurological diseases.
Collapse
Affiliation(s)
- J D Comer
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.,Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.,Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - S Alvarez
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - S J Butler
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - J A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
9
|
Duquette PM, Lamarche-Vane N. The calcium-activated protease calpain regulates netrin-1 receptor deleted in colorectal cancer-induced axon outgrowth in cortical neurons. J Neurochem 2019; 152:315-332. [PMID: 31344270 DOI: 10.1111/jnc.14837] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 07/02/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022]
Abstract
During development, neurons extend axons toward their appropriate synaptic targets to establish functional neuronal connections. The growth cone, a highly motile structure at the tip of the axon, is capable of recognizing extracellular guidance cues and translating them into directed axon outgrowth through modulation of the actin cytoskeleton. Netrin-1 mediates its attractive function through the receptor deleted in colorectal cancer (DCC) to promote axon outgrowth and guidance. The calcium-activated protease calpain is involved in the cleavage of cytoskeletal proteins, which plays an important role during adhesion turnover and cell migration. However, its function during neuronal development is less understood. Here we demonstrate that netrin-1 activated calpain in embryonic rat cortical neurons in an extracellular-regulated kinase 1/2-dependent manner. In addition, we found that netrin-1 stimulation led to an increase in calpain-1 localization in the axon, whereas its endogenous inhibitor calpastatin was decreased in the growth cones of cortical neurons by indirect immunofluorescence. Interestingly, calpain-1 was able to cleave DCC in vitro. Furthermore, netrin-1 induced the cleavage of the cytoskeletal proteins spectrin and focal adhesion kinase concomitantly with the intracellular domain of DCC in a calpain-dependent manner in embryonic rat cortical neurons. Cortical neurons over-expressing calpastatin or calpain-depleted neurons displayed increased basal axon length and were unresponsive to netrin-1 stimulation. Altogether, we propose a novel model whereby netrin-1/DCC-mediated axon outgrowth is modulated by calpain-mediated proteolysis of DCC and cytoskeletal targets in embryonic cortical neurons. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Philippe M Duquette
- Cancer Research Program, Research Institute of the McGill University Health Center (RI-MUHC), Montréal, Québec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Nathalie Lamarche-Vane
- Cancer Research Program, Research Institute of the McGill University Health Center (RI-MUHC), Montréal, Québec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| |
Collapse
|
10
|
Tulloch AJ, Teo S, Carvajal BV, Tessier-Lavigne M, Jaworski A. Diverse spinal commissural neuron populations revealed by fate mapping and molecular profiling using a novel Robo3 Cre mouse. J Comp Neurol 2019; 527:2948-2972. [PMID: 31152445 DOI: 10.1002/cne.24720] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/22/2019] [Accepted: 05/21/2019] [Indexed: 12/19/2022]
Abstract
The two sides of the nervous system coordinate and integrate information via commissural neurons, which project axons across the midline. Commissural neurons in the spinal cord are a highly heterogeneous population of cells with respect to their birthplace, final cell body position, axonal trajectory, and neurotransmitter phenotype. Although commissural axon guidance during development has been studied in great detail, neither the developmental origins nor the mature phenotypes of commissural neurons have been characterized comprehensively, largely due to lack of selective genetic access to these neurons. Here, we generated mice expressing Cre recombinase from the Robo3 locus specifically in commissural neurons. We used Robo3 Cre mice to characterize the transcriptome and various origins of developing commissural neurons, revealing new details about their extensive heterogeneity in molecular makeup and developmental lineage. Further, we followed the fate of commissural neurons into adulthood, thereby elucidating their settling positions and molecular diversity and providing evidence for possible functions in various spinal cord circuits. Our studies establish an important genetic entry point for further analyses of commissural neuron development, connectivity, and function.
Collapse
Affiliation(s)
- Alastair J Tulloch
- Department of Neuroscience, Brown University, Providence, Rhode Island.,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, Rhode Island
| | - Shaun Teo
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York
| | | | - Marc Tessier-Lavigne
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York.,Department of Biology, Stanford University, Stanford, California
| | - Alexander Jaworski
- Department of Neuroscience, Brown University, Providence, Rhode Island.,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, Rhode Island
| |
Collapse
|
11
|
Gorla M, Santiago C, Chaudhari K, Layman AAK, Oliver PM, Bashaw GJ. Ndfip Proteins Target Robo Receptors for Degradation and Allow Commissural Axons to Cross the Midline in the Developing Spinal Cord. Cell Rep 2019; 26:3298-3312.e4. [PMID: 30893602 PMCID: PMC6913780 DOI: 10.1016/j.celrep.2019.02.080] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 01/29/2019] [Accepted: 02/20/2019] [Indexed: 12/27/2022] Open
Abstract
Commissural axons initially respond to attractive signals at the midline, but once they cross, they become sensitive to repulsive cues. This switch prevents axons from re-entering the midline. In insects and mammals, negative regulation of Roundabout (Robo) receptors prevents premature response to the midline repellant Slit. In Drosophila, the endosomal protein Commissureless (Comm) prevents Robo1 surface expression before midline crossing by diverting Robo1 into late endosomes. Notably, Comm is not conserved in vertebrates. We identified two Nedd-4-interacting proteins, Ndfip1 and Ndfip2, that act analogously to Comm to localize Robo1 to endosomes. Ndfip proteins recruit Nedd4 E3 ubiquitin ligases to promote Robo1 ubiquitylation and degradation. Ndfip proteins are expressed in commissural axons in the developing spinal cord and removal of Ndfip proteins results in increased Robo1 expression and reduced midline crossing. Our results define a conserved Robo1 intracellular sorting mechanism between flies and mammals to avoid premature responsiveness to Slit.
Collapse
Affiliation(s)
- Madhavi Gorla
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Celine Santiago
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karina Chaudhari
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Awo Akosua Kesewa Layman
- The Children's Hospital of Philadelphia, Division of Protective Immunity, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Paula M Oliver
- The Children's Hospital of Philadelphia, Division of Protective Immunity, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Bonanomi D, Valenza F, Chivatakarn O, Sternfeld MJ, Driscoll SP, Aslanian A, Lettieri K, Gullo M, Badaloni A, Lewcock JW, Hunter T, Pfaff SL. p190RhoGAP Filters Competing Signals to Resolve Axon Guidance Conflicts. Neuron 2019; 102:602-620.e9. [PMID: 30902550 PMCID: PMC8608148 DOI: 10.1016/j.neuron.2019.02.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/05/2018] [Accepted: 02/19/2019] [Indexed: 12/21/2022]
Abstract
The rich functional diversity of the nervous system is founded in the specific connectivity of the underlying neural circuitry. Neurons are often preprogrammed to respond to multiple axon guidance signals because they use sequential guideposts along their pathways, but this necessitates a strict spatiotemporal regulation of intracellular signaling to ensure the cues are detected in the correct order. We performed a mouse mutagenesis screen and identified the Rho GTPase antagonist p190RhoGAP as a critical regulator of motor axon guidance. Rather than acting as a compulsory signal relay, p190RhoGAP uses a non-conventional GAP-independent mode to transiently suppress attraction to Netrin-1 while motor axons exit the spinal cord. Once in the periphery, a subset of axons requires p190RhoGAP-mediated inhibition of Rho signaling to target specific muscles. Thus, the multifunctional activity of p190RhoGAP emerges from its modular design. Our findings reveal a cell-intrinsic gate that filters conflicting signals, establishing temporal windows of signal detection.
Collapse
Affiliation(s)
- Dario Bonanomi
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA; San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 60, 20132 Milan, Italy.
| | - Fabiola Valenza
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 60, 20132 Milan, Italy
| | - Onanong Chivatakarn
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Matthew J Sternfeld
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Shawn P Driscoll
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Aaron Aslanian
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Karen Lettieri
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Miriam Gullo
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Aurora Badaloni
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 60, 20132 Milan, Italy
| | - Joseph W Lewcock
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Samuel L Pfaff
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Ryu JR, Kim JH, Cho HM, Jo Y, Lee B, Joo S, Chae U, Nam Y, Cho IJ, Sun W. A monitoring system for axonal growth dynamics using micropatterns of permissive and Semaphorin 3F chemorepulsive signals. LAB ON A CHIP 2019; 19:291-305. [PMID: 30539180 DOI: 10.1039/c8lc00845k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Neurons reach their correct targets by directional outgrowth of axons, which is mediated by attractive or repulsive cues. Growing axons occasionally cross a field of repulsive cues and stop at intermediate targets on the journey to their final destination. However, it is not well-understood how individual growth cones make decisions, and pass through repulsive territory to reach their permissive target regions. We developed a microcontact printing culture system that could trap individual axonal tips in a permissive dot area surrounded by the repulsive signal, semaphorin 3F (Sema3F). Axons of rat hippocampal neurons on the Sema3F/PLL dot array extended in the checkboard pattern with a significantly slow growth rate. The detailed analysis of the behaviors of axonal growth cones revealed the saccadic dynamics in the dot array system. The trapped axonal tips in the permissive area underwent growth cone enlargement with remarkably spiky filopodia, promoting their escape from the Sema3F constraints with straight extension of axons. This structured axonal growth on the dot pattern was disrupted by increased inter-dot distance, or perturbing intracellular signaling machineries. These data indicate that axons grow against repulsive signals by jumping over the repulsive cues, depending on contact signals and intracellular milieu. Our study suggests that our dot array culture system can be used as a screening system to easily and efficiently evaluate ECM or small molecule inhibitors interfering growth cone dynamics leading to controlling axonal growth.
Collapse
Affiliation(s)
- Jae Ryun Ryu
- Department of Anatomy, Brain Korea 21, Korea University College of Medicine, Anam-Dong, Sungbuk-Gu, Seoul, 136-705, Republic of Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Vuong TA, Lee SJ, Leem YE, Lee JR, Bae GU, Kang JS. SGTb regulates a surface localization of a guidance receptor BOC to promote neurite outgrowth. Cell Signal 2019; 55:100-108. [PMID: 30639199 DOI: 10.1016/j.cellsig.2019.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/02/2019] [Accepted: 01/05/2019] [Indexed: 01/07/2023]
Abstract
Neuritogenesis is a critical event for neuronal differentiation and neuronal circuitry formation during neuronal development and regeneration. Our previous study revealed a critical role of a guidance receptor BOC in a neuronal differentiation and neurite outgrowth. However, regulatory mechanisms for BOC signaling pathway remain largely unexplored. In the current study, we have identified Small glutamine-rich tetratricopeptide repeat (TPR)-containing b (SGTb) as a BOC interacting protein through yeast two-hybrid screening. Like BOC, SGTb is highly expressed in brain and P19 embryonal carcinoma (EC) cells differentiated into neuronal cells. BOC and SGTb proteins co-precipitate in mouse brain and differentiated P19 EC cells. Furthermore, BOC and SGTb co-localize in neurites and especially are concentrated at the tip of neurites in various neuronal cells. SGTb depletion attenuates neuronal differentiation of P19 cells through reduction of the surface level of BOC. Additionally, SGTb depletion causes BOC localization at neurite tip, coinciding with decreased p-JNK levels critical for actin cytoskeleton remodeling. The overexpression of SGTb or BOC restores JNK activation in BOC or SGTb-depleted cells, respectively. Finally, SGTb elevates the level of surface-resident BOC in BOC-depleted cells, restoring JNK activation. Taken together, our data suggest that SGTb interacts with BOC and regulates its surface level and consequent JNK activation, thereby promoting neuronal differentiation and neurite outgrowth.
Collapse
Affiliation(s)
- Tuan Anh Vuong
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Sang-Jin Lee
- Research Institute of Pharmaceutical Science, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Jae-Rin Lee
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Gyu-Un Bae
- Research Institute of Pharmaceutical Science, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea; Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Furlanis E, Scheiffele P. Regulation of Neuronal Differentiation, Function, and Plasticity by Alternative Splicing. Annu Rev Cell Dev Biol 2018; 34:451-469. [PMID: 30028642 DOI: 10.1146/annurev-cellbio-100617-062826] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Posttranscriptional mechanisms provide powerful means to expand the coding power of genomes. In nervous systems, alternative splicing has emerged as a fundamental mechanism not only for the diversification of protein isoforms but also for the spatiotemporal control of transcripts. Thus, alternative splicing programs play instructive roles in the development of neuronal cell type-specific properties, neuronal growth, self-recognition, synapse specification, and neuronal network function. Here we discuss the most recent genome-wide efforts on mapping RNA codes and RNA-binding proteins for neuronal alternative splicing regulation. We illustrate how alternative splicing shapes key steps of neuronal development, neuronal maturation, and synaptic properties. Finally, we highlight efforts to dissect the spatiotemporal dynamics of alternative splicing and their potential contribution to neuronal plasticity and the mature nervous system.
Collapse
|
16
|
Chen Z. Common cues wire the spinal cord: Axon guidance molecules in spinal neuron migration. Semin Cell Dev Biol 2018; 85:71-77. [PMID: 29274387 DOI: 10.1016/j.semcdb.2017.12.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 01/28/2023]
Abstract
Topographic arrangement of neuronal cell bodies and axonal tracts are crucial for proper wiring of the nervous system. This involves often-coordinated neuronal migration and axon guidance during development. Most neurons migrate from their birthplace to specific topographic coordinates as they adopt the final cell fates and extend axons. The axons follow temporospatial specific guidance cues to reach the appropriate targets. When neuronal or axonal migration or their coordination is disrupted, severe consequences including neurodevelopmental disorders and neurological diseases, can arise. Neuronal and axonal migration shares some molecular mechanisms, as genes originally identified as axon guidance molecules have been increasingly shown to direct both navigation processes. This review focuses on axon guidance pathways that are shown to also direct neuronal migration in the vertebrate spinal cord.
Collapse
Affiliation(s)
- Zhe Chen
- Department of MCD Biology, University of Colorado Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
17
|
Bonneaud N, Layalle S, Colomb S, Jourdan C, Ghysen A, Severac D, Dantec C, Nègre N, Maschat F. Control of nerve cord formation by Engrailed and Gooseberry-Neuro: A multi-step, coordinated process. Dev Biol 2017; 432:273-285. [PMID: 29097190 DOI: 10.1016/j.ydbio.2017.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 10/06/2017] [Accepted: 10/24/2017] [Indexed: 01/05/2023]
Abstract
One way to better understand the molecular mechanisms involved in the construction of a nervous system is to identify the downstream effectors of major regulatory proteins. We previously showed that Engrailed (EN) and Gooseberry-Neuro (GsbN) transcription factors act in partnership to drive the formation of posterior commissures in the central nervous system of Drosophila. In this report, we identified genes regulated by both EN and GsbN through chromatin immunoprecipitation ("ChIP on chip") and transcriptome experiments, combined to a genetic screen relied to the gene dose titration method. The genomic-scale approaches allowed us to define 175 potential targets of EN-GsbN regulation. We chose a subset of these genes to examine ventral nerve cord (VNC) defects and found that half of the mutated targets show clear VNC phenotypes when doubly heterozygous with en or gsbn mutations, or when homozygous. This strategy revealed new groups of genes never described for their implication in the construction of the nerve cord. Their identification suggests that, to construct the nerve cord, EN-GsbN may act at three levels, in: (i) sequential control of the attractive-repulsive signaling that ensures contralateral projection of the commissural axons, (ii) temporal control of the translation of some mRNAs, (iii) regulation of the capability of glial cells to act as commissural guideposts for developing axons. These results illustrate how an early, coordinated transcriptional control may orchestrate the various mechanisms involved in the formation of stereotyped neuronal networks. They also validate the overall strategy to identify genes that play crucial role in axonal pathfinding.
Collapse
Affiliation(s)
- Nathalie Bonneaud
- MMDN, Univ. Montpellier, EPHE, INSERM, U1198, Montpellier, F-34095 France; CNRS,UPR1142, Institut de Génétique Humaine, Montpellier, F-34094, France
| | - Sophie Layalle
- CNRS,UPR1142, Institut de Génétique Humaine, Montpellier, F-34094, France; CNRS - INSERM - Université de Montpellier, UMR-5203, Institut de Génomique Fonctionnelle, Montpellier F-34094, France
| | - Sophie Colomb
- CNRS,UPR1142, Institut de Génétique Humaine, Montpellier, F-34094, France
| | - Christophe Jourdan
- MMDN, Univ. Montpellier, EPHE, INSERM, U1198, Montpellier, F-34095 France
| | - Alain Ghysen
- MMDN, Univ. Montpellier, EPHE, INSERM, U1198, Montpellier, F-34095 France
| | - Dany Severac
- MGX - Montpellier GenomiX, Institut de Génomique Fonctionnelle, Montpellier F-34094, France
| | - Christelle Dantec
- MGX - Montpellier GenomiX, Institut de Génomique Fonctionnelle, Montpellier F-34094, France
| | - Nicolas Nègre
- DGIMI, INRA, Université de Montpellier, 34095 Montpellier, France; Institut Universitaire de France (IUF), Paris, France
| | - Florence Maschat
- MMDN, Univ. Montpellier, EPHE, INSERM, U1198, Montpellier, F-34095 France; CNRS,UPR1142, Institut de Génétique Humaine, Montpellier, F-34094, France.
| |
Collapse
|
18
|
Howard LJ, Brown HE, Wadsworth BC, Evans TA. Midline axon guidance in the Drosophila embryonic central nervous system. Semin Cell Dev Biol 2017; 85:13-25. [PMID: 29174915 DOI: 10.1016/j.semcdb.2017.11.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 02/02/2023]
Abstract
Studies in the fruit fly Drosophila melanogaster have provided many fundamental insights into the genetic regulation of neural development, including the identification and characterization of evolutionarily conserved axon guidance pathways and their roles in important guidance decisions. Due to its highly organized and fast-developing embryonic nervous system, relatively small number of neurons, and molecular and genetic tools for identifying, labeling, and manipulating individual neurons or small neuronal subsets, studies of axon guidance in the Drosophila embryonic CNS have allowed researchers to dissect these genetic mechanisms with a high degree of precision. In this review, we discuss the major axon guidance pathways that regulate midline crossing of axons and the formation and guidance of longitudinal axon tracts, two processes that contribute to the development of the precise three-dimensional structure of the insect nerve cord. We focus particularly on recent insights into the roles and regulation of canonical midline axon guidance pathways, and on additional factors and pathways that have recently been shown to contribute to axon guidance decisions at and near the midline.
Collapse
Affiliation(s)
- LaFreda J Howard
- Department of Biological Sciences, University of Arkansas, Fayetteville AR 72701, USA
| | - Haley E Brown
- Department of Biological Sciences, University of Arkansas, Fayetteville AR 72701, USA
| | - Benjamin C Wadsworth
- Department of Biological Sciences, University of Arkansas, Fayetteville AR 72701, USA
| | - Timothy A Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville AR 72701, USA.
| |
Collapse
|
19
|
Protein O-Mannosyltransferases Affect Sensory Axon Wiring and Dynamic Chirality of Body Posture in the Drosophila Embryo. J Neurosci 2017; 38:1850-1865. [PMID: 29167399 DOI: 10.1523/jneurosci.0346-17.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 11/01/2017] [Accepted: 11/06/2017] [Indexed: 02/06/2023] Open
Abstract
Genetic defects in protein O-mannosyltransferase 1 (POMT1) and POMT2 underlie severe muscular dystrophies. POMT genes are evolutionarily conserved in metazoan organisms. In Drosophila, both male and female POMT mutants show a clockwise rotation of adult abdominal segments, suggesting a chirality of underlying pathogenic mechanisms. Here we described and analyzed a similar phenotype in POMT mutant embryos that shows left-handed body torsion. Our experiments demonstrated that coordinated muscle contraction waves are associated with asymmetric embryo rolling, unveiling a new chirality marker in Drosophila development. Using genetic and live-imaging approaches, we revealed that the torsion phenotype results from differential rolling and aberrant patterning of peristaltic waves of muscle contractions. Our results demonstrated that peripheral sensory neurons are required for normal contractions that prevent the accumulation of torsion. We found that POMT mutants show abnormal axonal connections of sensory neurons. POMT transgenic expression limited to sensory neurons significantly rescued the torsion phenotype, axonal connectivity defects, and abnormal contractions in POMT mutant embryos. Together, our data suggested that protein O-mannosylation is required for normal sensory feedback to control coordinated muscle contractions and body posture. This mechanism may shed light on analogous functions of POMT genes in mammals and help to elucidate the etiology of neurological defects in muscular dystrophies.SIGNIFICANCE STATEMENT Protein O-mannosyltransferases (POMTs) are evolutionarily conserved in metazoans. Mutations in POMTs cause severe muscular dystrophies associated with pronounced neurological defects. However, neurological functions of POMTs remain poorly understood. We demonstrated that POMT mutations in Drosophila result in abnormal muscle contractions and cause embryo torsion. Our experiments uncovered a chirality of embryo movements and a unique POMT-dependent mechanism that maintains symmetry of a developing system affected by chiral forces. Furthermore, POMTs were found to be required for proper axon connectivity of sensory neurons, suggesting that O-mannosylation regulates the sensory feedback controlling muscle contractions. This novel POMT function in the peripheral nervous system may shed light on analogous functions in mammals and help to elucidate pathomechanisms of neurological abnormalities in muscular dystrophies.
Collapse
|
20
|
Abstract
Motor neurons of the spinal cord are responsible for the assembly of neuromuscular connections indispensable for basic locomotion and skilled movements. A precise spatial relationship exists between the position of motor neuron cell bodies in the spinal cord and the course of their axonal projections to peripheral muscle targets. Motor neuron innervation of the vertebrate limb is a prime example of this topographic organization and by virtue of its accessibility and predictability has provided access to fundamental principles of motor system development and neuronal guidance. The seemingly basic binary map established by genetically defined motor neuron subtypes that target muscles in the limb is directed by a surprisingly large number of directional cues. Rather than being simply redundant, these converging signaling pathways are hierarchically linked and cooperate to increase the fidelity of axon pathfinding decisions. A current priority is to determine how multiple guidance signals are integrated by individual growth cones and how they synergize to delineate class-specific axonal trajectories.
Collapse
Affiliation(s)
- Dario Bonanomi
- Molecular Neurobiology Laboratory, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
21
|
Marsh APL, Edwards TJ, Galea C, Cooper HM, Engle EC, Jamuar SS, Méneret A, Moutard ML, Nava C, Rastetter A, Robinson G, Rouleau G, Roze E, Spencer-Smith M, Trouillard O, Billette de Villemeur T, Walsh CA, Yu TW, Heron D, Sherr EH, Richards LJ, Depienne C, Leventer RJ, Lockhart PJ. DCC mutation update: Congenital mirror movements, isolated agenesis of the corpus callosum, and developmental split brain syndrome. Hum Mutat 2017; 39:23-39. [PMID: 29068161 DOI: 10.1002/humu.23361] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 10/08/2017] [Accepted: 10/11/2017] [Indexed: 12/12/2022]
Abstract
The deleted in colorectal cancer (DCC) gene encodes the netrin-1 (NTN1) receptor DCC, a transmembrane protein required for the guidance of commissural axons. Germline DCC mutations disrupt the development of predominantly commissural tracts in the central nervous system (CNS) and cause a spectrum of neurological disorders. Monoallelic, missense, and predicted loss-of-function DCC mutations cause congenital mirror movements, isolated agenesis of the corpus callosum (ACC), or both. Biallelic, predicted loss-of-function DCC mutations cause developmental split brain syndrome (DSBS). Although the underlying molecular mechanisms leading to disease remain poorly understood, they are thought to stem from reduced or perturbed NTN1 signaling. Here, we review the 26 reported DCC mutations associated with abnormal CNS development in humans, including 14 missense and 12 predicted loss-of-function mutations, and discuss their associated clinical characteristics and diagnostic features. We provide an update on the observed genotype-phenotype relationships of congenital mirror movements, isolated ACC and DSBS, and correlate this to our current understanding of the biological function of DCC in the development of the CNS. All mutations and their associated phenotypes were deposited into a locus-specific LOVD (https://databases.lovd.nl/shared/genes/DCC).
Collapse
Affiliation(s)
- Ashley P L Marsh
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Timothy J Edwards
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Australia.,Faculty of Medicine, The University of Queensland, Herston, Brisbane, Australia
| | - Charles Galea
- Drug Delivery, Disposition and Dynamics (D4), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Australia
| | - Elizabeth C Engle
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts.,Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts.,Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts.,Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts
| | - Saumya S Jamuar
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts.,Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts.,Department of Paediatrics, KK Women's and Children's Hospital, Paediatric Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Aurélie Méneret
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Département de Neurologie, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Marie-Laure Moutard
- Service de Neuropédiatrie, AP-HP, Hôpital Trousseau, Paris, France.,UPMC, GRC ConCer-LD, Sorbonne Université, Paris, France.,Centre de référence "Neurogénétique", Paris, France
| | - Caroline Nava
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Département de Génétique, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Agnès Rastetter
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Gail Robinson
- Neuropsychology Research Unit, School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Guy Rouleau
- Department of Neurology and Neurosurgery, McGill University Health Center, Montreal, Quebec, Canada.,Montreal Neurological Institute and Hospital, McGill University, Montréal, Quebec, Canada
| | - Emmanuel Roze
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Département de Neurologie, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Megan Spencer-Smith
- Clinical Sciences, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Clayton Campus, Clayton, Victoria, Australia
| | - Oriane Trouillard
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Thierry Billette de Villemeur
- Service de Neuropédiatrie, AP-HP, Hôpital Trousseau, Paris, France.,UPMC, GRC ConCer-LD, Sorbonne Université, Paris, France.,Centre de Référence "déficiences intellectuelles de causes rares", Paris, France.,INSERM U1141, Paris, France
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts.,Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts.,Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts.,Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Timothy W Yu
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | | | - Delphine Heron
- UPMC, GRC ConCer-LD, Sorbonne Université, Paris, France.,Département de Génétique, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Elliott H Sherr
- Department of Neurology, UCSF Benioff Children's Hospital, San Francisco, California
| | - Linda J Richards
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Australia.,The University of Queensland, School of Biomedical Sciences, St Lucia, Brisbane, Australia
| | - Christel Depienne
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Département de Génétique, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France.,Département de Médicine translationnelle et Neurogénétique, IGBMC, CNRS UMR 7104, INSERM U964, Université de Strasbourg, Illkirch, France.,Laboratoires de génétique, Institut de génétique médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Richard J Leventer
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Neuroscience Research Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Neurology, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Paul J Lockhart
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
22
|
Gamboa NT, Taussky P, Park MS, Couldwell WT, Mahan MA, Kalani MYS. Neurovascular patterning cues and implications for central and peripheral neurological disease. Surg Neurol Int 2017; 8:208. [PMID: 28966815 PMCID: PMC5609400 DOI: 10.4103/sni.sni_475_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 06/28/2017] [Indexed: 12/20/2022] Open
Abstract
The highly branched nervous and vascular systems run along parallel trajectories throughout the human body. This stereotyped pattern of branching shared by the nervous and vascular systems stems from a common reliance on specific cues critical to both neurogenesis and angiogenesis. Continually emerging evidence supports the notion of later-evolving vascular networks co-opting neural molecular mechanisms to ensure close proximity and adequate delivery of oxygen and nutrients to nervous tissue. As our understanding of these biologic pathways and their phenotypic manifestations continues to advance, identification of where pathways go awry will provide critical insight into central and peripheral nervous system pathology.
Collapse
Affiliation(s)
- Nicholas T Gamboa
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Philipp Taussky
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Min S Park
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - William T Couldwell
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Mark A Mahan
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - M Yashar S Kalani
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
23
|
Banerjee S, Mino RE, Fisher ES, Bhat MA. A versatile genetic tool to study midline glia function in the Drosophila CNS. Dev Biol 2017; 429:35-43. [PMID: 28602954 PMCID: PMC5554714 DOI: 10.1016/j.ydbio.2017.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/01/2017] [Accepted: 06/08/2017] [Indexed: 11/30/2022]
Abstract
Neuron-glial interactions are crucial for growth, guidance and ensheathment of axons across species. In the Drosophila CNS midline, neuron-glial interactions underlie ensheathment of commissural axons by midline glial (MG) cells in a manner similar to mammalian oligodendrocytes. Although there has been some advance in the study of neuron-glial interactions and ensheathment of axons in the CNS midline, key aspects of axonal ensheathment are still not fully understood. One of the limitations has been the unavailability of MG membrane markers that could highlight the glial processes wrapping the axons. Previous studies have identified two key molecular players from the neuronal and glial cell types in the CNS midline. These are the neuronal transmembrane protein Neurexin IV (Nrx IV) and the membrane-anchored MG protein Wrapper, both of which interact in trans to mediate neuron-glial interactions and ensheathment of commissural axons. In the current study, we attempt to further our understanding of MG biology and try to overcome some of the technical difficulties posed by the lack of a robust MG driver that will specifically allow expression or knockdown of genes in MG. We report the generation of BAC transgenic flies of wrapper-GAL4 and demonstrate how these flies could be used as a genetic tool to understand MG biology. We have utilized the GAL4/UAS system to drive GFP-reporter lines (membrane-bound mCD8-GFP; microtubule-associated tau-GFP) and nuclear lacZ using wrapper-GAL4 to highlight the MG cells and/or their processes that surround and perform axonal ensheathment functions in the embryonic midline. We also describe the utility of the wrapper-GAL4 driver line to down-regulate known MG genes specifically in Wrapper-positive cells. Finally, we validate the functionality of the wrapper-GAL4 driver by rescue of wrapper mutant phenotypes and lethality. Together, these studies provide us with a versatile genetic tool to investigate MG functions and will aid in future investigations where genetic screens using wrapper-GAL4 could be designed to identify novel molecular players at the Drosophila midline and unravel key aspects of MG biology.
Collapse
Affiliation(s)
- Swati Banerjee
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| | - Rosa E Mino
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Elizabeth S Fisher
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| |
Collapse
|
24
|
Schaefer N, Rotermund C, Blumrich EM, Lourenco MV, Joshi P, Hegemann RU, Jamwal S, Ali N, García Romero EM, Sharma S, Ghosh S, Sinha JK, Loke H, Jain V, Lepeta K, Salamian A, Sharma M, Golpich M, Nawrotek K, Paidi RK, Shahidzadeh SM, Piermartiri T, Amini E, Pastor V, Wilson Y, Adeniyi PA, Datusalia AK, Vafadari B, Saini V, Suárez-Pozos E, Kushwah N, Fontanet P, Turner AJ. The malleable brain: plasticity of neural circuits and behavior - a review from students to students. J Neurochem 2017. [PMID: 28632905 DOI: 10.1111/jnc.14107] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One of the most intriguing features of the brain is its ability to be malleable, allowing it to adapt continually to changes in the environment. Specific neuronal activity patterns drive long-lasting increases or decreases in the strength of synaptic connections, referred to as long-term potentiation and long-term depression, respectively. Such phenomena have been described in a variety of model organisms, which are used to study molecular, structural, and functional aspects of synaptic plasticity. This review originated from the first International Society for Neurochemistry (ISN) and Journal of Neurochemistry (JNC) Flagship School held in Alpbach, Austria (Sep 2016), and will use its curriculum and discussions as a framework to review some of the current knowledge in the field of synaptic plasticity. First, we describe the role of plasticity during development and the persistent changes of neural circuitry occurring when sensory input is altered during critical developmental stages. We then outline the signaling cascades resulting in the synthesis of new plasticity-related proteins, which ultimately enable sustained changes in synaptic strength. Going beyond the traditional understanding of synaptic plasticity conceptualized by long-term potentiation and long-term depression, we discuss system-wide modifications and recently unveiled homeostatic mechanisms, such as synaptic scaling. Finally, we describe the neural circuits and synaptic plasticity mechanisms driving associative memory and motor learning. Evidence summarized in this review provides a current view of synaptic plasticity in its various forms, offers new insights into the underlying mechanisms and behavioral relevance, and provides directions for future research in the field of synaptic plasticity. Read the Editorial Highlight for this article on page 788. Cover Image for this issue: doi: 10.1111/jnc.13815.
Collapse
Affiliation(s)
- Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University of Wuerzburg, Würzburg, Germany
| | - Carola Rotermund
- German Center of Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Eva-Maria Blumrich
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany.,Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pooja Joshi
- Inserm UMR 1141, Robert Debre Hospital, Paris, France
| | - Regina U Hegemann
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Sumit Jamwal
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Nilufar Ali
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | | | - Sorabh Sharma
- Neuropharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Shampa Ghosh
- National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Tarnaka, Hyderabad, India
| | - Jitendra K Sinha
- National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Tarnaka, Hyderabad, India
| | - Hannah Loke
- Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Vishal Jain
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Katarzyna Lepeta
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ahmad Salamian
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mahima Sharma
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mojtaba Golpich
- Department of Medicine, University Kebangsaan Malaysia Medical Centre (HUKM), Cheras, Kuala Lumpur, Malaysia
| | - Katarzyna Nawrotek
- Department of Process Thermodynamics, Faculty of Process and Environmental Engineering, Lodz University of Technology, Lodz, Poland
| | - Ramesh K Paidi
- CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sheila M Shahidzadeh
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York, USA
| | - Tetsade Piermartiri
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Elham Amini
- Department of Medicine, University Kebangsaan Malaysia Medical Centre (HUKM), Cheras, Kuala Lumpur, Malaysia
| | - Veronica Pastor
- Instituto de Biología Celular y Neurociencia Prof. Eduardo De Robertis, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Yvette Wilson
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Philip A Adeniyi
- Cell Biology and Neurotoxicity Unit, Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado - Ekiti, Ekiti State, Nigeria
| | | | - Benham Vafadari
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Vedangana Saini
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Edna Suárez-Pozos
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Toxicología, México
| | - Neetu Kushwah
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Paula Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cellular Biology and Neuroscience (IBCN), CONICET-UBA, School of Medicine, Buenos Aires, Argentina
| | - Anthony J Turner
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
25
|
Suter TACS, DeLoughery ZJ, Jaworski A. Meninges-derived cues control axon guidance. Dev Biol 2017; 430:1-10. [PMID: 28784295 DOI: 10.1016/j.ydbio.2017.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 01/10/2023]
Abstract
The axons of developing neurons travel long distances along stereotyped pathways under the direction of extracellular cues sensed by the axonal growth cone. Guidance cues are either secreted proteins that diffuse freely or bind the extracellular matrix, or membrane-anchored proteins. Different populations of axons express distinct sets of receptors for guidance cues, which results in differential responses to specific ligands. The full repertoire of axon guidance cues and receptors and the identity of the tissues producing these cues remain to be elucidated. The meninges are connective tissue layers enveloping the vertebrate brain and spinal cord that serve to protect the central nervous system (CNS). The meninges also instruct nervous system development by regulating the generation and migration of neural progenitors, but it has not been determined whether they help guide axons to their targets. Here, we investigate a possible role for the meninges in neuronal wiring. Using mouse neural tissue explants, we show that developing spinal cord meninges produce secreted attractive and repulsive cues that can guide multiple types of axons in vitro. We find that motor and sensory neurons, which project axons across the CNS-peripheral nervous system (PNS) boundary, are attracted by meninges. Conversely, axons of both ipsi- and contralaterally projecting dorsal spinal cord interneurons are repelled by meninges. The responses of these axonal populations to the meninges are consistent with their trajectories relative to meninges in vivo, suggesting that meningeal guidance factors contribute to nervous system wiring and control which axons are able to traverse the CNS-PNS boundary.
Collapse
Affiliation(s)
- Tracey A C S Suter
- Department of Neuroscience, Brown University, Providence, RI 02912, United States
| | - Zachary J DeLoughery
- Department of Neuroscience, Brown University, Providence, RI 02912, United States
| | - Alexander Jaworski
- Department of Neuroscience, Brown University, Providence, RI 02912, United States.
| |
Collapse
|
26
|
Early Commissural Diencephalic Neurons Control Habenular Axon Extension and Targeting. Curr Biol 2017; 27:270-278. [PMID: 28065605 DOI: 10.1016/j.cub.2016.11.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/07/2016] [Accepted: 11/16/2016] [Indexed: 01/19/2023]
Abstract
Most neuronal populations form on both the left and right sides of the brain. Their efferent axons appear to grow synchronously along similar pathways on each side, although the neurons or their environment often differ between the two hemispheres [1-4]. How this coordination is controlled has received little attention. Frequently, neurons establish interhemispheric connections, which can function to integrate information between brain hemispheres (e.g., [5]). Such commissures form very early, suggesting their potential developmental role in coordinating ipsilateral axon navigation during embryonic development [4]. To address the temporal-spatial control of bilateral axon growth, we applied long-term time-lapse imaging to visualize the formation of the conserved left-right asymmetric habenular neural circuit in the developing zebrafish embryo [6]. Although habenular neurons are born at different times across brain hemispheres [7], we found that elongation of habenular axons occurs synchronously. The initiation of axon extension is not controlled within the habenular network itself but through an early developing proximal diencephalic network. The commissural neurons of this network influence habenular axons both ipsilaterally and contralaterally. Their unilateral absence impairs commissure formation and coordinated habenular axon elongation and causes their subsequent arrest on both sides of the brain. Thus, habenular neural circuit formation depends on a second intersecting commissural network, which facilitates the exchange of information between hemispheres required for ipsilaterally projecting habenular axons. This mechanism of network formation may well apply to other circuits, and has only remained undiscovered due to technical limitations.
Collapse
|
27
|
Oliva C, Hassan BA. Receptor Tyrosine Kinases and Phosphatases in Neuronal Wiring: Insights From Drosophila. Curr Top Dev Biol 2016; 123:399-432. [PMID: 28236973 DOI: 10.1016/bs.ctdb.2016.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tyrosine phosphorylation is at the crossroads of many signaling pathways. Brain wiring is not an exception, and several receptor tyrosine kinases (RTKs) and tyrosine receptor phosphates (RPTPs) have been involved in this process. Considerable work has been done on RTKs, and for many of them, detailed molecular mechanisms and functions in several systems have been characterized. In contrast, RPTPs have been studied considerably less and little is known about their ligands and substrates. In both families, we find redundancy between different members to accomplish particular wiring patterns. Strikingly, some RTKs and RPTPs have lost their catalytic activity during evolution, but not their importance in biological processes. In this regard, we have to keep in mind that these proteins have multiple domains and some of their functions are independent of tyrosine phosphorylation/dephosphorylation. Since RTKs and RPTPs are enzymes involved not only in early stages of axon and dendrite pathfinding but also in synapse formation and physiology, they have a potential as drug targets. Drosophila has been a key model organism in the search of a better understanding of brain wiring, and its sophisticated toolbox is very suitable for studying the function of genes with pleiotropic functions such as RTKs and RPTPs, from wiring to synaptic formation and function. In these review, we mainly cover findings from this model organism and complement them with discoveries in vertebrate systems.
Collapse
Affiliation(s)
- Carlos Oliva
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad of Chile, Santiago, Chile.
| | - Bassem A Hassan
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, AP-HP, Institut du Cerveau et la Moelle (ICM)-Hôpital Pitié-Salpêtrière, Boulevard de l'Hôpital, Paris, France.
| |
Collapse
|
28
|
Vuong TA, Leem YE, Kim BG, Cho H, Lee SJ, Bae GU, Kang JS. A Sonic hedgehog coreceptor, BOC regulates neuronal differentiation and neurite outgrowth via interaction with ABL and JNK activation. Cell Signal 2016; 30:30-40. [PMID: 27871935 DOI: 10.1016/j.cellsig.2016.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022]
Abstract
Neurite outgrowth is a critical step for neurogenesis and remodeling synaptic circuitry during neuronal development and regeneration. An immunoglobulin superfamily member, BOC functions as Sonic hedgehog (Shh) coreceptor in canonical and noncanonical Shh signaling in neuronal development and axon outgrowth/guidance. However signaling mechanisms responsible for BOC action during these processes remain unknown. In our previous studies, a multiprotein complex containing BOC and a closely related protein CDO promotes myogenic differentiation through activation of multiple signaling pathways, including non-receptor tyrosine kinase ABL. Given that ABL and Jun. N-terminal kinase (JNK) are implicated in actin cytoskeletal dynamics required for neurogenesis, we investigated the relationship between BOC, ABL and JNK during neuronal differentiation. Here, we demonstrate that BOC and ABL are induced in P19 embryonal carcinoma (EC) cells and cortical neural progenitor cells (NPCs) during neuronal differentiation. BOC-depleted EC cells or Boc-/- NPCs exhibit impaired neuronal differentiation with shorter neurite formation. BOC interacts with ABL through its putative SH2 binding domain and seems to be phosphorylated in an ABL activity-dependent manner. Unlike wildtype BOC, ABL-binding defective BOC mutants exhibit impaired JNK activation and neuronal differentiation. Finally, Shh treatment enhances JNK activation which is diminished by BOC depletion. These data suggest that BOC interacts with ABL and activates JNK thereby promoting neuronal differentiation and neurite outgrowth.
Collapse
Affiliation(s)
- Tuan Anh Vuong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Bok-Geon Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Sang-Jin Lee
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Gyu-Un Bae
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea.
| |
Collapse
|
29
|
Chisholm AD, Hutter H, Jin Y, Wadsworth WG. The Genetics of Axon Guidance and Axon Regeneration in Caenorhabditis elegans. Genetics 2016; 204:849-882. [PMID: 28114100 PMCID: PMC5105865 DOI: 10.1534/genetics.115.186262] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/06/2016] [Indexed: 11/18/2022] Open
Abstract
The correct wiring of neuronal circuits depends on outgrowth and guidance of neuronal processes during development. In the past two decades, great progress has been made in understanding the molecular basis of axon outgrowth and guidance. Genetic analysis in Caenorhabditis elegans has played a key role in elucidating conserved pathways regulating axon guidance, including Netrin signaling, the slit Slit/Robo pathway, Wnt signaling, and others. Axon guidance factors were first identified by screens for mutations affecting animal behavior, and by direct visual screens for axon guidance defects. Genetic analysis of these pathways has revealed the complex and combinatorial nature of guidance cues, and has delineated how cues guide growth cones via receptor activity and cytoskeletal rearrangement. Several axon guidance pathways also affect directed migrations of non-neuronal cells in C. elegans, with implications for normal and pathological cell migrations in situations such as tumor metastasis. The small number of neurons and highly stereotyped axonal architecture of the C. elegans nervous system allow analysis of axon guidance at the level of single identified axons, and permit in vivo tests of prevailing models of axon guidance. C. elegans axons also have a robust capacity to undergo regenerative regrowth after precise laser injury (axotomy). Although such axon regrowth shares some similarities with developmental axon outgrowth, screens for regrowth mutants have revealed regeneration-specific pathways and factors that were not identified in developmental screens. Several areas remain poorly understood, including how major axon tracts are formed in the embryo, and the function of axon regeneration in the natural environment.
Collapse
Affiliation(s)
| | - Harald Hutter
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, V5A 1S6, Canada
| | - Yishi Jin
- Section of Neurobiology, Division of Biological Sciences, and
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093
- Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, Chevy Chase, Maryland, and
| | - William G Wadsworth
- Department of Pathology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| |
Collapse
|
30
|
Junge HJ, Yung AR, Goodrich LV, Chen Z. Netrin1/DCC signaling promotes neuronal migration in the dorsal spinal cord. Neural Dev 2016; 11:19. [PMID: 27784329 PMCID: PMC5081974 DOI: 10.1186/s13064-016-0074-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/17/2016] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Newborn neurons often migrate before undergoing final differentiation, extending neurites, and forming synaptic connections. Therefore, neuronal migration is crucial for establishing neural circuitry during development. In the developing spinal cord, neuroprogenitors first undergo radial migration within the ventricular zone. Differentiated neurons continue to migrate tangentially before reaching the final positions. The molecular pathways that regulate these migration processes remain largely unknown. Our previous study suggests that the DCC receptor is important for the migration of the dorsal spinal cord progenitors and interneurons. In this study, we determined the involvement of the Netrin1 ligand and the ROBO3 coreceptor in the migration. RESULTS By pulse labeling neuroprogenitors with electroporation, we examined their radial migration in Netrin1 (Ntn1), Dcc, and Robo3 knockout mice. We found that all three mutants exhibit delayed migration. Furthermore, using immunohistochemistry of the BARHL2 interneuron marker, we found that the mediolateral and dorsoventral migration of differentiated dorsal interneurons is also delayed. Together, our results suggest that Netrin1/DCC signaling induce neuronal migration in the dorsal spinal cord. CONCLUSIONS Netrin1, DCC, and ROBO3 have been extensively studied for their functions in regulating axon guidance in the spinal commissural interneurons. We reveal that during earlier development of dorsal interneurons including commissural neurons, these molecules play an important role in promoting cell migration.
Collapse
Affiliation(s)
- Harald J Junge
- Department of MCDB, University of Colorado, Boulder, CO, 80309, USA.
| | - Andrea R Yung
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Zhe Chen
- Department of MCDB, University of Colorado, Boulder, CO, 80309, USA.
| |
Collapse
|
31
|
Stanic K, Saldivia N, Förstera B, Torrejón M, Montecinos H, Caprile T. Expression Patterns of Extracellular Matrix Proteins during Posterior Commissure Development. Front Neuroanat 2016; 10:89. [PMID: 27733818 PMCID: PMC5039192 DOI: 10.3389/fnana.2016.00089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/12/2016] [Indexed: 11/13/2022] Open
Abstract
Extracellular matrix (ECM) molecules are pivotal for central nervous system (CNS) development, facilitating cell migration, axonal growth, myelination, dendritic spine formation, and synaptic plasticity, among other processes. During axon guidance, the ECM not only acts as a permissive or non-permissive substrate for navigating axons, but also modulates the effects of classical guidance cues, such as netrin or Eph/ephrin family members. Despite being highly important, little is known about the expression of ECM molecules during CNS development. Therefore, this study assessed the molecular expression patterns of tenascin, HNK-1, laminin, fibronectin, perlecan, decorin, and osteopontin along chick embryo prosomere 1 during posterior commissure development. The posterior commissure is the first transversal axonal tract of the embryonic vertebrate brain. Located in the dorso-caudal portion of prosomere 1, posterior commissure axons primarily arise from the neurons of basal pretectal nuclei that run dorsally to the roof plate midline, where some turn toward the ipsilateral side. Expressional analysis of ECM molecules in this area these revealed to be highly arranged, and molecule interactions with axon fascicles suggested involvement in processes other than structural support. In particular, tenascin and the HNK-1 epitope extended in ventro-dorsal columns and enclosed axons during navigation to the roof plate. Laminin and osteopontin were expressed in the midline, very close to axons that at this point must decide between extending to the contralateral side or turning to the ipsilateral side. Finally, fibronectin, decorin, and perlecan appeared unrelated to axonal pathfinding in this region and were instead restricted to the external limiting membrane. In summary, the present report provides evidence for an intricate expression of different extracellular molecules that may cooperate in guiding posterior commissure axons.
Collapse
Affiliation(s)
- Karen Stanic
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Natalia Saldivia
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Benjamín Förstera
- Department of Physiology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Marcela Torrejón
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Hernán Montecinos
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Teresa Caprile
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| |
Collapse
|
32
|
Yakimova AO, Pugacheva OM, Golubkova EV, Mamon LA. Cytoplasmic localization of SBR (Dm NXF1) protein and its zonal distribution in the ganglia of Drosophila melanogaster larvae. INVERTEBRATE NEUROSCIENCE 2016; 16:9. [DOI: 10.1007/s10158-016-0192-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 06/24/2016] [Indexed: 10/21/2022]
|
33
|
Preitner N, Quan J, Li X, Nielsen FC, Flanagan JG. IMP2 axonal localization, RNA interactome, and function in the development of axon trajectories. Development 2016; 143:2753-9. [PMID: 27385015 DOI: 10.1242/dev.128348] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/13/2016] [Indexed: 01/28/2023]
Abstract
RNA-based regulatory mechanisms play important roles in the development and plasticity of neural circuits and neurological disease. Developing axons provide a model well suited to the study of RNA-based regulation, and contain specific subsets of mRNAs that are locally translated and have roles in axon pathfinding. However, the RNA-binding proteins involved in axon pathfinding, and their corresponding mRNA targets, are still largely unknown. Here we find that the RNA-binding protein IMP2 (Igf2bp2) is strikingly enriched in developing axon tracts, including in spinal commissural axons. We used the HITS-CLIP approach to perform a genome-wide identification of RNAs that interact directly with IMP2 in the native context of developing mouse brain. This IMP2 interactome was highly enriched for mRNA targets related to axon guidance. Accordingly, IMP2 knockdown in the developing spinal cord led to strong defects in commissural axon trajectories at the midline intermediate target. These results reveal a highly distinctive axonal enrichment of IMP2, show that it interacts with a network of axon guidance-related mRNAs, and reveal that it is required for normal axon pathfinding during vertebrate development.
Collapse
Affiliation(s)
- Nicolas Preitner
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Jie Quan
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Xinmin Li
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Finn C Nielsen
- Center for Genomic Medicine, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, Copenhagen Ø DK-2100, Denmark
| | - John G Flanagan
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
34
|
Leggere JC, Saito Y, Darnell RB, Tessier-Lavigne M, Junge HJ, Chen Z. NOVA regulates Dcc alternative splicing during neuronal migration and axon guidance in the spinal cord. eLife 2016; 5. [PMID: 27223328 PMCID: PMC4930329 DOI: 10.7554/elife.14264] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/23/2016] [Indexed: 02/03/2023] Open
Abstract
RNA-binding proteins (RBPs) control multiple aspects of post-transcriptional gene regulation and function during various biological processes in the nervous system. To further reveal the functional significance of RBPs during neural development, we carried out an in vivo RNAi screen in the dorsal spinal cord interneurons, including the commissural neurons. We found that the NOVA family of RBPs play a key role in neuronal migration, axon outgrowth, and axon guidance. Interestingly, Nova mutants display similar defects as the knockout of the Dcc transmembrane receptor. We show here that Nova deficiency disrupts the alternative splicing of Dcc, and that restoring Dcc splicing in Nova knockouts is able to rescue the defects. Together, our results demonstrate that the production of DCC splice variants controlled by NOVA has a crucial function during many stages of commissural neuron development.
Collapse
Affiliation(s)
- Janelle C Leggere
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, United States
| | - Yuhki Saito
- Laboratory of Molecular Neuro-Oncology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Robert B Darnell
- Laboratory of Molecular Neuro-Oncology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Marc Tessier-Lavigne
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, United States
| | - Harald J Junge
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, United States
| | - Zhe Chen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, United States
| |
Collapse
|
35
|
Myosin Va and Endoplasmic Reticulum Calcium Channel Complex Regulates Membrane Export during Axon Guidance. Cell Rep 2016; 15:1329-44. [DOI: 10.1016/j.celrep.2016.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 03/11/2016] [Accepted: 03/31/2016] [Indexed: 11/22/2022] Open
|
36
|
Noraz N, Jaaoini I, Charoy C, Watrin C, Chounlamountri N, Benon A, Malleval C, Boudin H, Honnorat J, Castellani V, Pellier-Monnin V. Syk kinases are required for spinal commissural axon repulsion at the midline via the ephrin/Eph pathway. Development 2016; 143:2183-93. [PMID: 27122172 DOI: 10.1242/dev.128629] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 04/15/2016] [Indexed: 12/26/2022]
Abstract
In the hematopoietic system, Syk family tyrosine kinases are essential components of immunoreceptor ITAM-based signaling. While there is increasing data indicating the involvement of immunoreceptors in neural functions, the contribution of Syk kinases remains obscure. Previously, we identified phosphorylated forms of Syk kinases in specialized populations of migrating neurons or projecting axons. Moreover, we identified ephrin/Eph as guidance molecules utilizing the ITAM-bearing CD3zeta (Cd247) and associated Syk kinases for the growth cone collapse response induced in vitro Here, we show that in the developing spinal cord, Syk is phosphorylated in navigating commissural axons. By analyzing axon trajectories in open-book preparations of Syk(-/-); Zap70(-/-) mouse embryos, we show that Syk kinases are dispensable for attraction towards the midline but confer growth cone responsiveness to repulsive signals that expel commissural axons from the midline. Known to serve a repulsive function at the midline, ephrin B3/EphB2 are obvious candidates for driving the Syk-dependent repulsive response. Indeed, Syk kinases were found to be required for ephrin B3-induced growth cone collapse in cultured commissural neurons. In fragments of commissural neuron-enriched tissues, Syk is in a constitutively phosphorylated state and ephrin B3 decreased its level of phosphorylation. Direct pharmacological inhibition of Syk kinase activity was sufficient to induce growth cone collapse. In conclusion, Syk kinases act as a molecular switch of growth cone adhesive and repulsive responses.
Collapse
Affiliation(s)
- Nelly Noraz
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Iness Jaaoini
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Camille Charoy
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Chantal Watrin
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Naura Chounlamountri
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Aurélien Benon
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Céline Malleval
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Hélène Boudin
- INSERM U1064, Institut de Transplantation Urologie-Néphrologie, Nantes F-44035, France
| | - Jérôme Honnorat
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France Hospices Civils de Lyon, Lyon F-69000, France
| | - Valérie Castellani
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Véronique Pellier-Monnin
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| |
Collapse
|
37
|
Akita T, Kumada T, Yoshihara SI, Egea J, Yamagishi S. Ion channels, guidance molecules, intracellular signaling and transcription factors regulating nervous and vascular system development. J Physiol Sci 2016; 66:175-88. [PMID: 26507418 PMCID: PMC4752580 DOI: 10.1007/s12576-015-0416-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 01/13/2023]
Abstract
Our sophisticated thoughts and behaviors are based on the miraculous development of our complex nervous network system, in which many different types of proteins and signaling cascades are regulated in a temporally and spatially ordered manner. Here we review our recent attempts to grasp the principles of nervous system development in terms of general cellular phenomena and molecules, such as volume-regulated anion channels, intracellular Ca(2+) and cyclic nucleotide signaling, the Npas4 transcription factor and the FLRT family of axon guidance molecules. We also present an example illustrating that the same FLRT family may regulate the development of vascular networks as well. The aim of this review is to open up new vistas for understanding the intricacy of nervous and vascular system development.
Collapse
Affiliation(s)
- Tenpei Akita
- Department of Neurophysiology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan.
| | - Tatsuro Kumada
- Department of Occupational Therapy, Faculty of Health and Medical Sciences, Tokoha University, 1230 Miyakoda-cho, Kita-ku, Hamamatsu, Shizuoka, 431-2102, Japan
| | - Sei-ichi Yoshihara
- Laboratory for Molecular Biology of Neural System, Advanced Medical Research Center, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| | - Joaquim Egea
- Molecular and Developmental Neurobiology Group, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida, 25198, Lleida, Spain
| | - Satoru Yamagishi
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| |
Collapse
|
38
|
Cate MS, Gajendra S, Alsbury S, Raabe T, Tear G, Mitchell KJ. Mushroom body defect is required in parallel to Netrin for midline axon guidance in Drosophila. Development 2016; 143:972-7. [PMID: 26893348 PMCID: PMC4813287 DOI: 10.1242/dev.129684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 01/29/2016] [Indexed: 12/23/2022]
Abstract
The outgrowth of many neurons within the central nervous system is initially directed towards or away from the cells lying at the midline. Recent genetic evidence suggests that a simple model of differential sensitivity to the conserved Netrin attractants and Slit repellents is insufficient to explain the guidance of all axons at the midline. In the Drosophila embryonic ventral nerve cord, many axons still cross the midline in the absence of the Netrin genes (NetA and NetB) or their receptor frazzled. Here we show that mutation of mushroom body defect (mud) dramatically enhances the phenotype of Netrin or frazzled mutants, resulting in many more axons failing to cross the midline, although mutations in mud alone have little effect. This suggests that mud, which encodes a microtubule-binding coiled-coil protein homologous to NuMA and LIN-5, is an essential component of a Netrin-independent pathway that acts in parallel to promote midline crossing. We demonstrate that this novel role of Mud in axon guidance is independent of its previously described role in neural precursor development. These studies identify a parallel pathway controlling midline guidance in Drosophila and highlight a novel role for Mud potentially acting downstream of Frizzled to aid axon guidance.
Collapse
Affiliation(s)
- Marie-Sophie Cate
- MRC Centre for Developmental Neurobiology, New Hunts House, King's College, London, SE1 1UL, UK
| | - Sangeetha Gajendra
- MRC Centre for Developmental Neurobiology, New Hunts House, King's College, London, SE1 1UL, UK
| | - Samantha Alsbury
- MRC Centre for Developmental Neurobiology, New Hunts House, King's College, London, SE1 1UL, UK
| | - Thomas Raabe
- MSZ Universitat Würzburg, Versbacher Strasse 5, Würzberg 97078, Germany
| | - Guy Tear
- MRC Centre for Developmental Neurobiology, New Hunts House, King's College, London, SE1 1UL, UK
| | - Kevin J Mitchell
- Howard Hughes Medical Institute, Division of Neurobiology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA Smurfit Institute of Genetics and Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
39
|
Frizzled3 Controls Axonal Polarity and Intermediate Target Entry during Striatal Pathway Development. J Neurosci 2016; 35:14205-19. [PMID: 26490861 DOI: 10.1523/jneurosci.1840-15.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED The striatum is a large brain nucleus with an important role in the control of movement and emotions. Medium spiny neurons (MSNs) are striatal output neurons forming prominent descending axon tracts that target different brain nuclei. However, how MSN axon tracts in the forebrain develop remains poorly understood. Here, we implicate the Wnt binding receptor Frizzled3 in several uncharacterized aspects of MSN pathway formation [i.e., anterior-posterior guidance of MSN axons in the striatum and their subsequent growth into the globus pallidus (GP), an important (intermediate) target]. In Frizzled3 knock-out mice, MSN axons fail to extend along the anterior-posterior axis of the striatum, and many do not reach the GP. Wnt5a acts as an attractant for MSN axons in vitro, is expressed in a posterior high, anterior low gradient in the striatum, and Wnt5a knock-out mice phenocopy striatal anterior-posterior defects observed in Frizzled3 mutants. This suggests that Wnt5a controls anterior-posterior guidance of MSN axons through Frizzled3. Axons that reach the GP in Frizzled3 knock-out mice fail to enter this structure. Surprisingly, entry of MSN axons into the GP non-cell-autonomously requires Frizzled3, and our data suggest that GP entry may be contingent on the correct positioning of "corridor" guidepost cells for thalamocortical axons by Frizzled3. Together, these data dissect MSN pathway development and reveal (non)cell-autonomous roles for Frizzled3 in MSN axon guidance. Further, they are the first to identify a gene that provides anterior-posterior axon guidance in a large brain nucleus and link Frizzled3 to corridor cell development. SIGNIFICANCE STATEMENT Striatal axon pathways mediate complex physiological functions and are an important therapeutic target, underscoring the need to define how these connections are established. Remarkably, the molecular programs regulating striatal pathway development remain poorly characterized. Here, we determine the embryonic ontogeny of the two main striatal pathways (striatonigral and striatopallidal) and identify novel (non)cell-autonomous roles for the axon guidance receptor Frizzled3 in uncharacterized aspects of striatal pathway formation (i.e., anterior-posterior axon guidance in the striatum and axon entry into the globus pallidus). Further, our results link Frizzled3 to corridor guidepost cell development and suggest that an abnormal distribution of these cells has unexpected, widespread effects on the development of different axon tracts (i.e., striatal and thalamocortical axons).
Collapse
|
40
|
Schiweck J, Beauchamp M, Humo M, Lelievre V. Old friends, new story: The role of Slit2C signaling through PlexinA1. Cell Adh Migr 2015; 9:417-21. [PMID: 26632339 DOI: 10.1080/19336918.2015.1106670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Growth cone guidance is driven by attractive and repulsive signaling cues. Until recently, repulsive signaling by semaphorins was thought to be mediated through Plexin receptors, whereas Slits-induced repulsion was solely mediated through Robo receptors. In a recent report published in Nature Neuroscience, Celine Delloye-Bourgeois and colleagues (2015) combined phenotypic analyses of transgenic mouse lines and in vitro biochemical experiments to identify PlexinA1 as a novel receptor for Slits. Strikingly, they uncovered for the very first time that the Slit2C-terminal fragment possesses some unique biological activity as binding partner for PlexinA1. Even more excitingly, the signaling cascade triggered by SlitC binding to PlexinA1 mediates growth cone collapse of commissural axons both in vivo and ex vivo and nicely complements Robo-Slit signaling in the developing spinal cord midline to prevent midline recrossing.
Collapse
Affiliation(s)
- Juliane Schiweck
- a Joint Master in Neuroscience; University of Strasbourg-France ; Strasbourg , France
| | - Marta Beauchamp
- a Joint Master in Neuroscience; University of Strasbourg-France ; Strasbourg , France
| | - Muris Humo
- a Joint Master in Neuroscience; University of Strasbourg-France ; Strasbourg , France
| | - Vincent Lelievre
- a Joint Master in Neuroscience; University of Strasbourg-France ; Strasbourg , France
| |
Collapse
|
41
|
Jaworski A, Tom I, Tong RK, Gildea HK, Koch AW, Gonzalez LC, Tessier-Lavigne M. Operational redundancy in axon guidance through the multifunctional receptor Robo3 and its ligand NELL2. Science 2015; 350:961-5. [PMID: 26586761 DOI: 10.1126/science.aad2615] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Axon pathfinding is orchestrated by numerous guidance cues, including Slits and their Robo receptors, but it remains unclear how information from multiple cues is integrated or filtered. Robo3, a Robo family member, allows commissural axons to reach and cross the spinal cord midline by antagonizing Robo1/2-mediated repulsion from midline-expressed Slits and potentiating deleted in colorectal cancer (DCC)-mediated midline attraction to Netrin-1, but without binding either Slits or Netrins. We identified a secreted Robo3 ligand, neural epidermal growth factor-like-like 2 (NELL2), which repels mouse commissural axons through Robo3 and helps steer them to the midline. These findings identify NELL2 as an axon guidance cue and establish Robo3 as a multifunctional regulator of pathfinding that simultaneously mediates NELL2 repulsion, inhibits Slit repulsion, and facilitates Netrin attraction to achieve a common guidance purpose.
Collapse
Affiliation(s)
- Alexander Jaworski
- Division of Research, Genentech, South San Francisco, CA 94080, USA. Laboratory of Brain Development and Repair, The Rockefeller University, New York, NY 10065, USA. Department of Neuroscience, Brown University, Providence, RI 02912, USA.
| | - Irene Tom
- Department of Protein Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Raymond K Tong
- Department of Protein Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Holly K Gildea
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Alexander W Koch
- Department of Protein Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Lino C Gonzalez
- Department of Protein Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Marc Tessier-Lavigne
- Division of Research, Genentech, South San Francisco, CA 94080, USA. Laboratory of Brain Development and Repair, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
42
|
Araújo SJ. The Hedgehog Signalling Pathway in Cell Migration and Guidance: What We Have Learned from Drosophila melanogaster. Cancers (Basel) 2015; 7:2012-22. [PMID: 26445062 PMCID: PMC4695873 DOI: 10.3390/cancers7040873] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/23/2015] [Accepted: 09/28/2015] [Indexed: 01/20/2023] Open
Abstract
Cell migration and guidance are complex processes required for morphogenesis, the formation of tumor metastases, and the progression of human cancer. During migration, guidance molecules induce cell directionality and movement through complex intracellular mechanisms. Expression of these molecules has to be tightly regulated and their signals properly interpreted by the receiving cells so as to ensure correct navigation. This molecular control is fundamental for both normal morphogenesis and human disease. The Hedgehog (Hh) signaling pathway is evolutionarily conserved and known to be crucial for normal cellular growth and differentiation throughout the animal kingdom. The relevance of Hh signaling for human disease is emphasized by its activation in many cancers. Here, I review the current knowledge regarding the involvement of the Hh pathway in cell migration and guidance during Drosophila development and discuss its implications for human cancer origin and progression.
Collapse
Affiliation(s)
- Sofia J Araújo
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Parc Cientific de Barcelona, C. Baldiri Reixac 10,08028 Barcelona, Spain.
| |
Collapse
|
43
|
Hara S, Kaneyama T, Inamata Y, Onodera R, Shirasaki R. Interstitial branch formation within the red nucleus by deep cerebellar nuclei-derived commissural axons during target recognition. J Comp Neurol 2015; 524:999-1014. [DOI: 10.1002/cne.23888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/29/2015] [Accepted: 08/21/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Satoshi Hara
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences; Osaka University; Suita Osaka 565-0871 Japan
| | - Takeshi Kaneyama
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences; Osaka University; Suita Osaka 565-0871 Japan
| | - Yasuyuki Inamata
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences; Osaka University; Suita Osaka 565-0871 Japan
| | - Ryota Onodera
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences; Osaka University; Suita Osaka 565-0871 Japan
| | - Ryuichi Shirasaki
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences; Osaka University; Suita Osaka 565-0871 Japan
| |
Collapse
|
44
|
Blasiak A, Lee GU, Kilinc D. Neuron Subpopulations with Different Elongation Rates and DCC Dynamics Exhibit Distinct Responses to Isolated Netrin-1 Treatment. ACS Chem Neurosci 2015; 6:1578-90. [PMID: 26190161 DOI: 10.1021/acschemneuro.5b00142] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Correct wiring of the nervous system requires guidance cues, diffusible or substrate-bound proteins that steer elongating axons to their target tissues. Netrin-1, the best characterized member of the Netrins family of guidance molecules, is known to induce axon turning and modulate axon elongation rate; however, the factors regulating the axonal response to Netrin-1 are not fully understood. Using microfluidics, we treated fluidically isolated axons of mouse primary cortical neurons with Netrin-1 and characterized axon elongation rates, as well as the membrane localization of deleted in colorectal cancer (DCC), a well-established receptor of Netrin-1. The capacity to stimulate and observe a large number of individual axons allowed us to conduct distribution analyses, through which we identified two distinct neuron subpopulations based on different elongation behavior and different DCC membrane dynamics. Netrin-1 reduced the elongation rates in both subpopulations, where the effect was more pronounced in the slow growing subpopulation. Both the source of Ca(2+) influx and the basal cytosolic Ca(2+) levels regulated the effect of Netrin-1, for example, Ca(2+) efflux from the endoplasmic reticulum due to the activation of Ryanodine channels blocked Netrin-1-induced axon slowdown. Netrin-1 treatment resulted in a rapid membrane insertion of DCC, followed by a gradual internalization. DCC membrane dynamics were different in the central regions of the growth cones compared to filopodia and axon shafts, highlighting the temporal and spatial heterogeneity in the signaling events downstream of Netrin-1. Cumulatively, these results demonstrate the power of microfluidic compartmentalization and distribution analysis in describing the complex axonal Netrin-1 response.
Collapse
Affiliation(s)
- Agata Blasiak
- Bionanoscience Group, School
of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin
4, Ireland
| | - Gil U. Lee
- Bionanoscience Group, School
of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin
4, Ireland
| | - Devrim Kilinc
- Bionanoscience Group, School
of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin
4, Ireland
| |
Collapse
|
45
|
Comer JD, Pan FC, Willet SG, Haldipur P, Millen KJ, Wright CVE, Kaltschmidt JA. Sensory and spinal inhibitory dorsal midline crossing is independent of Robo3. Front Neural Circuits 2015; 9:36. [PMID: 26257608 PMCID: PMC4511845 DOI: 10.3389/fncir.2015.00036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/02/2015] [Indexed: 11/25/2022] Open
Abstract
Commissural neurons project across the midline at all levels of the central nervous system (CNS), providing bilateral communication critical for the coordination of motor activity and sensory perception. Midline crossing at the spinal ventral midline has been extensively studied and has revealed that multiple developmental lineages contribute to this commissural neuron population. Ventral midline crossing occurs in a manner dependent on Robo3 regulation of Robo/Slit signaling and the ventral commissure is absent in the spinal cord and hindbrain of Robo3 mutants. Midline crossing in the spinal cord is not limited to the ventral midline, however. While prior anatomical studies provide evidence that commissural axons also cross the midline dorsally, little is known of the genetic and molecular properties of dorsally-crossing neurons or of the mechanisms that regulate dorsal midline crossing. In this study, we describe a commissural neuron population that crosses the spinal dorsal midline during the last quarter of embryogenesis in discrete fiber bundles present throughout the rostrocaudal extent of the spinal cord. Using immunohistochemistry, neurotracing, and mouse genetics, we show that this commissural neuron population includes spinal inhibitory neurons and sensory nociceptors. While the floor plate and roof plate are dispensable for dorsal midline crossing, we show that this population depends on Robo/Slit signaling yet crosses the dorsal midline in a Robo3-independent manner. The dorsally-crossing commissural neuron population we describe suggests a substrate circuitry for pain processing in the dorsal spinal cord.
Collapse
Affiliation(s)
- John D Comer
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences New York, NY, USA ; Developmental Biology Program, Sloan-Kettering Institute New York, NY, USA ; Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program New York, NY, USA
| | - Fong Cheng Pan
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Spencer G Willet
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Parthiv Haldipur
- Seattle Children's Research Institute, Center for Integrative Brain Research Seattle, WA, USA
| | - Kathleen J Millen
- Seattle Children's Research Institute, Center for Integrative Brain Research Seattle, WA, USA ; Department of Pediatrics, Genetics Division, University of Washington Seattle, WA, USA
| | - Christopher V E Wright
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Julia A Kaltschmidt
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences New York, NY, USA ; Developmental Biology Program, Sloan-Kettering Institute New York, NY, USA
| |
Collapse
|
46
|
Regulation of gene expression through production of unstable mRNA isoforms. Biochem Soc Trans 2015; 42:1196-205. [PMID: 25110025 DOI: 10.1042/bst20140102] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alternative splicing is universally accredited for expanding the information encoded within the transcriptome. In recent years, several tightly regulated alternative splicing events have been reported which do not lead to generation of protein products, but lead to unstable mRNA isoforms. Instead these transcripts are targets for NMD (nonsense-mediated decay) or retained in the nucleus and degraded. In the present review I discuss the regulation of these events, and how many have been implicated in control of gene expression that is instrumental to a number of developmental paradigms. I further discuss their relevance to disease settings and conclude by highlighting technologies that will aid identification of more candidate events in future.
Collapse
|
47
|
Butler SJ, Bronner ME. From classical to current: analyzing peripheral nervous system and spinal cord lineage and fate. Dev Biol 2015; 398:135-46. [PMID: 25446276 PMCID: PMC4845735 DOI: 10.1016/j.ydbio.2014.09.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/22/2014] [Accepted: 09/25/2014] [Indexed: 01/13/2023]
Abstract
During vertebrate development, the central (CNS) and peripheral nervous systems (PNS) arise from the neural plate. Cells at the margin of the neural plate give rise to neural crest cells, which migrate extensively throughout the embryo, contributing to the majority of neurons and all of the glia of the PNS. The rest of the neural plate invaginates to form the neural tube, which expands to form the brain and spinal cord. The emergence of molecular cloning techniques and identification of fluorophores like Green Fluorescent Protein (GFP), together with transgenic and electroporation technologies, have made it possible to easily visualize the cellular and molecular events in play during nervous system formation. These lineage-tracing techniques have precisely demonstrated the migratory pathways followed by neural crest cells and increased knowledge about their differentiation into PNS derivatives. Similarly, in the spinal cord, lineage-tracing techniques have led to a greater understanding of the regional organization of multiple classes of neural progenitor and post-mitotic neurons along the different axes of the spinal cord and how these distinct classes of neurons assemble into the specific neural circuits required to realize their various functions. Here, we review how both classical and modern lineage and marker analyses have expanded our knowledge of early peripheral nervous system and spinal cord development.
Collapse
Affiliation(s)
- Samantha J Butler
- Department of Neurobiology, TLSB 3129, 610 Charles E Young Drive East, University of California, Los Angeles, Los Angeles, CA 90095-7239, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Marianne E Bronner
- Department of Neurobiology, TLSB 3129, 610 Charles E Young Drive East, University of California, Los Angeles, Los Angeles, CA 90095-7239, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
48
|
Santiago C, Bashaw GJ. Transcription factors and effectors that regulate neuronal morphology. Development 2015; 141:4667-80. [PMID: 25468936 DOI: 10.1242/dev.110817] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Transcription factors establish the tremendous diversity of cell types in the nervous system by regulating the expression of genes that give a cell its morphological and functional properties. Although many studies have identified requirements for specific transcription factors during the different steps of neural circuit assembly, few have identified the downstream effectors by which they control neuronal morphology. In this Review, we highlight recent work that has elucidated the functional relationships between transcription factors and the downstream effectors through which they regulate neural connectivity in multiple model systems, with a focus on axon guidance and dendrite morphogenesis.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Abstract
Recent findings in several organ systems show that cytoneme-mediated signaling transports signaling proteins along cellular extensions and targets cell-to-cell exchanges to synaptic contacts. This mechanism of paracrine signaling may be a general one that is used by many (or all) cell types in many (or all) organs. We briefly review these findings in this perspective. We also describe the properties of several signaling systems that have previously been interpreted to support a passive diffusion mechanism of signaling protein dispersion, but can now be understood in the context of the cytoneme mechanism. Also watch the Video Abstract.
Collapse
Affiliation(s)
- Sougata Roy
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Thomas B. Kornberg
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
50
|
Organisti C, Hein I, Grunwald Kadow IC, Suzuki T. Flamingo, a seven-pass transmembrane cadherin, cooperates with Netrin/Frazzled in Drosophila midline guidance. Genes Cells 2014; 20:50-67. [PMID: 25440577 DOI: 10.1111/gtc.12202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 10/01/2014] [Indexed: 01/31/2023]
Abstract
During central nervous system development, several guidance cues and receptors, as well as cell adhesion molecules, are required for guiding axons across the midline and along the anterior-posterior axis. In Drosophila, commissural axons sense the midline attractants Netrin A and B (Net) through Frazzled (Fra) receptors. Despite their importance, lack of Net or fra affects only some commissures, suggesting that additional molecules can fulfill this function. Recently, planar cell polarity (PCP) proteins have been implicated in midline axon guidance in both vertebrate and invertebrate systems. Here, we report that the atypical cadherin and PCP molecule Flamingo/Starry night (Fmi/Stan) acts jointly with Net/Fra signaling during midline development. Additional removal of fmi strongly increases the guidance defects in Net/fra mutants. Rescue and domain deletion experiments suggest that Fmi signaling facilitates commissural pathfinding potentially by mediating axonal fasciculation in a partly homophilic manner. Altogether, our results indicate that contact-mediated cell adhesion via Fmi acts in addition to the Net/Fra guidance system during axon pathfinding across the midline, underlining the importance of PCP molecules during vertebrates and invertebrates midline development.
Collapse
Affiliation(s)
- Cristina Organisti
- Max Planck Institute of Neurobiology, Sensory Neurogenetics Research Group, Am Klopferspitz 18, Martinsried, 82152, Germany
| | | | | | | |
Collapse
|