1
|
Clancy S, Xie N, Eluvathingal Muttikkal T, Wang J, Adylkhan A, Fateh E, Smith M, Wilson P, Smith M, Hogan A, Sutherland A, Lu X. Rac1 and Nectin3 are essential for planar cell polarity-directed axon guidance in the peripheral auditory system. Development 2025; 152:dev204423. [PMID: 40207531 PMCID: PMC12045628 DOI: 10.1242/dev.204423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/12/2025] [Indexed: 04/11/2025]
Abstract
Spiral ganglion neurons (SGNs) carry sound information from the cochlea to the hindbrain, and innervate either inner or outer hair cells. Type II SGNs (SGNIIs) extend peripheral afferents towards outer hair cells, which make a characteristic 90° turn towards the cochlear base and innervate multiple outer hair cells. It has been shown that the planar cell polarity (PCP) pathway acts non-autonomously in the cochlear epithelium to guide SGNII peripheral afferent turning. However, the underlying mechanisms are unknown. Here, we show that PCP signaling regulates junctional localization of the small GTPase Rac1 and the cell adhesion molecule Nectin3 in mouse cochlear supporting cells (SCs), which serve as intermediate targets of SGNII peripheral afferents. Loss of Rac1 or Nectin3 partially phenocopied SGNII peripheral afferent turning defects in Vangl2 mutants. We present evidence that Rac1 plays a non-autonomous role in part by regulating the localization of core PCP proteins Vangl2 and Dvl3 at the SC-SC junctions, while Nectin3 likely serves a cell adhesion function to control SGNII afferent turning. Together, these experiments identify Nectin3 and Rac1 as regulators of PCP-directed SGNII axon guidance in the cochlea.
Collapse
Affiliation(s)
- Shaylyn Clancy
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Nicholas Xie
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | | | - Jasmine Wang
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Aray Adylkhan
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Esha Fateh
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Margaret Smith
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Phillip Wilson
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Matthew Smith
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Arielle Hogan
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Ann Sutherland
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Xiaowei Lu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22903, USA
| |
Collapse
|
2
|
Neuman K, Zhang X, Lejeune BT, Pizzarella D, Vázquez M, Lewis LH, Koppes AN, Koppes RA. Static Magnetic Stimulation and Magnetic Microwires Synergistically Enhance and Guide Neurite Outgrowth. Adv Healthc Mater 2025; 14:e2403956. [PMID: 39568232 PMCID: PMC11773108 DOI: 10.1002/adhm.202403956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Indexed: 11/22/2024]
Abstract
Axonal growth is heavily influenced by topography and biophysical stimuli including magnetic and electrical fields. Despite extensive investigation, the degree of influence and the underlying genetic mechanisms remain poorly understood. Here, a novel approach to guide neurite growth is undertaken using an innovative ferromagnetic composite material - glass-coated magnetic microwire - to furnish a synergistic combination of magnetic and topographical cues. Whole rat dorsal root ganglia (DRG) are cultured under five different conditions: control, static magnetic field, magnetic microwire, static magnetic field + glass fiber, and static magnetic field + magnetic microwire. DRG outgrowth responses under each condition, including total neurite outgrowth and directionality, are compared. The combination of both magnetic stimulation and topography significantly increases total neurite outgrowth compared to the controls. The combination of magnetic stimulation and magnetic microwire lead to a strong directional bias of growth along the microwire, double what is observed with the glass fiber. Next generation RNA sequencing of DRG exposed to static magnetic field + magnetic microwire reveals the downregulation of genes relating to the immune response, interleukin signaling, and signal transduction. These results set the stage for contemplating future biophysical stimulation for axonal guidance and improved understanding of material-tissue interactions.
Collapse
Affiliation(s)
- Katelyn Neuman
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | - Xiaoyu Zhang
- Dept. of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Brian. T. Lejeune
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | | | - Manuel Vázquez
- Instituto de Ciencia de Materiales de MadridCSICMadrid28049Spain
| | - Laura H. Lewis
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
- Dept. of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Abigail N. Koppes
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
- Dept. of BioengineeringNortheastern UniversityBostonMA02115USA
- Dept. of BiologyNortheastern UniversityBostonMA02115USA
| | - Ryan A. Koppes
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| |
Collapse
|
3
|
Cammarata GM, Erdogan B, Sabo J, Kayaer Y, Dujava Zdimalova M, Engström F, Gupta U, Senel J, O'Brien T, Sibanda C, Thawani A, Folker ES, Braun M, Lansky Z, Lowery LA. The TOG5 domain of CKAP5 is required to interact with F-actin and promote microtubule advancement in neurons. Mol Biol Cell 2024; 35:br24. [PMID: 39504455 PMCID: PMC11656482 DOI: 10.1091/mbc.e24-05-0202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Microtubule (MT) and F-actin cytoskeletal cross-talk and organization are important aspects of axon guidance mechanisms, but how associated proteins facilitate this function remains largely unknown. While the MT-associated protein, CKAP5 (XMAP215/ch-TOG), has been best characterized as a MT polymerase, we have recently highlighted a novel role for CKAP5 in facilitating interactions between MT and F-actin in vitro and in embryonic Xenopus laevis neuronal growth cones. However, the mechanism by which it does so is unclear. Here, using in vitro reconstitution assays coupled with total internal reflection fluorescence microscopy, we report that the TOG5 domain of CKAP5 is necessary for its ability to bind to and bundle actin filaments, as well as to cross-link MTs and F-actin in vitro. Additionally, we show that this novel MT/F-actin cross-linking function of CKAP5 is possible even in MT polymerase-incompetent mutants of CKAP5 in vivo. Indeed, CKAP5 requires both MT and F-actin binding, but not MT polymerization, to promote MT-F-actin alignment in growth cones and axon outgrowth. Taken together, our findings provide mechanistic insights into how MT populations penetrate the growth cone periphery through CKAP5-facilitated interaction with F-actin during axon outgrowth and guidance.
Collapse
Affiliation(s)
| | - Burcu Erdogan
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Jan Sabo
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic 25240
| | - Yusuf Kayaer
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Michaela Dujava Zdimalova
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic 25240
- Department of Cell Biology, Faculty of Science, Charles University, Prague 12800, Czech Republic
| | - Filip Engström
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Urvika Gupta
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Jasming Senel
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Tara O'Brien
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Chiedza Sibanda
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Akanksha Thawani
- Department of Molecular and Cell Biology, UC Berkeley, Berkeley CA 94720
| | - Eric S Folker
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | - Marcus Braun
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic 25240
| | - Zdenek Lansky
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic 25240
| | - Laura A Lowery
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| |
Collapse
|
4
|
Xiang Z, He S, Chen R, Liu S, Liu M, Xu L, Zheng J, Jiang Z, Ma L, Sun Y, Qin Y, Chen Y, Li W, Wang X, Chen G, Lei W. Two-photon live imaging of direct glia-to-neuron conversion in the mouse cortex. Neural Regen Res 2024; 19:1781-1788. [PMID: 38103245 PMCID: PMC10960291 DOI: 10.4103/1673-5374.386401] [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: 04/28/2023] [Revised: 08/23/2023] [Accepted: 09/26/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00032/figure1/v/2023-12-16T180322Z/r/image-tiff Over the past decade, a growing number of studies have reported transcription factor-based in situ reprogramming that can directly convert endogenous glial cells into functional neurons as an alternative approach for neuroregeneration in the adult mammalian central nervous system. However, many questions remain regarding how a terminally differentiated glial cell can transform into a delicate neuron that forms part of the intricate brain circuitry. In addition, concerns have recently been raised around the absence of astrocyte-to-neuron conversion in astrocytic lineage-tracing mice. In this study, we employed repetitive two-photon imaging to continuously capture the in situ astrocyte-to-neuron conversion process following ectopic expression of the neural transcription factor NeuroD1 in both proliferating reactive astrocytes and lineage-traced astrocytes in the mouse cortex. Time-lapse imaging over several weeks revealed the step-by-step transition from a typical astrocyte with numerous short, tapered branches to a typical neuron with a few long neurites and dynamic growth cones that actively explored the local environment. In addition, these lineage-converting cells were able to migrate radially or tangentially to relocate to suitable positions. Furthermore, two-photon Ca2+ imaging and patch-clamp recordings confirmed that the newly generated neurons exhibited synchronous calcium signals, repetitive action potentials, and spontaneous synaptic responses, suggesting that they had made functional synaptic connections within local neural circuits. In conclusion, we directly visualized the step-by-step lineage conversion process from astrocytes to functional neurons in vivo and unambiguously demonstrated that adult mammalian brains are highly plastic with respect to their potential for neuroregeneration and neural circuit reconstruction.
Collapse
Affiliation(s)
- Zongqin Xiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
- Laboratory for Neuroimmunology in Health and Diseases, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Shu He
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Rongjie Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Shanggong Liu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Minhui Liu
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Flemish Region, Belgium
| | - Liang Xu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Jiajun Zheng
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Zhouquan Jiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Long Ma
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Ying Sun
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Yongpeng Qin
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Yi Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Wen Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
5
|
Clancy S, Xie N, Muttikkal TE, Wang J, Fateh E, Smith M, Wilson P, Smith M, Hogan A, Sutherland A, Lu X. Rac1 and Nectin3 are essential for PCP-directed axon guidance in the peripheral auditory system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597585. [PMID: 38895287 PMCID: PMC11185701 DOI: 10.1101/2024.06.05.597585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Our sense of hearing is critically dependent on the spiral ganglion neurons (SGNs) that connect the sound receptors in the organ of Corti (OC) to the cochlear nuclei of the hindbrain. Type I SGNs innervate inner hair cells (IHCs) to transmit sound signals, while type II SGNs (SGNIIs) innervate outer hair cells (OHCs) to detect moderate-to-intense sound. During development, SGNII afferents make a characteristic 90-degree turn toward the base of the cochlea and innervate multiple OHCs. It has been shown that the Planar Cell Polarity (PCP) pathway acts non-autonomously to mediate environmental cues in the cochlear epithelium for SGNII afferent turning towards the base. However, the underlying mechanisms are unknown. Here, we present evidence that PCP signaling regulates multiple downstream effectors to influence cell adhesion and the cytoskeleton in cochlear supporting cells (SCs), which serve as intermediate targets of SGNII afferents. We show that the core PCP gene Vangl2 regulates the localization of the small GTPase Rac1 and the cell adhesion molecule Nectin3 at SC-SC junctions through which SGNII afferents travel. Through in vivo genetic analysis, we also show that loss of Rac1 or Nectin3 partially phenocopied SGNII peripheral afferent turning defects in Vangl2 mutants, and that Rac1 plays a non-autonomous role in this process in part by regulating PCP protein localization at the SC-SC junctions. Additionally, epistasis analysis indicates that Nectin3 and Rac1 likely act in the same genetic pathway to control SGNII afferent turning. Together, these experiments identify Nectin3 and Rac1 as novel regulators of PCP-directed SGNII axon guidance in the cochlea.
Collapse
Affiliation(s)
- Shaylyn Clancy
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | - Nicholas Xie
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | | | - Jasmine Wang
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | - Esha Fateh
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | - Margaret Smith
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | - Phillip Wilson
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | - Matthew Smith
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | - Arielle Hogan
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | - Ann Sutherland
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| | - Xiaowei Lu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22903, United States
| |
Collapse
|
6
|
Iwanaga R, Yahagi N, Hakeda‐Suzuki S, Suzuki T. Cell adhesion and actin dynamics factors promote axonal extension and synapse formation in transplanted Drosophila photoreceptor cells. Dev Growth Differ 2024; 66:205-218. [PMID: 38403285 PMCID: PMC11457513 DOI: 10.1111/dgd.12916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/27/2024]
Abstract
Vision is formed by the transmission of light stimuli to the brain through axons extending from photoreceptor cells. Damage to these axons leads to loss of vision. Despite research on neural circuit regeneration through transplantation, achieving precise axon projection remains challenging. To achieve optic nerve regeneration by transplantation, we employed the Drosophila visual system. We previously established a transplantation method for Drosophila utilizing photoreceptor precursor cells extracted from the eye disc. However, little axonal elongation of transplanted cells into the brain, the lamina, was observed. We verified axonal elongation to the lamina by modifying the selection process for transplanted cells. Moreover, we focused on N-cadherin (Ncad), a cell adhesion factor, and Twinstar (Tsr), which has been shown to promote actin reorganization and induce axon elongation in damaged nerves. Overexpression of Ncad and tsr promoted axon elongation to the lamina, along with presynaptic structure formation in the elongating axons. Furthermore, overexpression of Neurexin-1 (Nrx-1), encoding a protein identified as a synaptic organizer, was found to not only promote presynapse formation but also enhance axon elongation. By introducing Ncad, tsr, and Nrx-1, we not only successfully achieved axonal projection of transplanted cells to the brain beyond the retina, but also confirmed the projection of transplanted cells into a deeper ganglion, the medulla. The present study offers valuable insights to realize regeneration through transplantation in a more complex nervous system.
Collapse
Affiliation(s)
- Riku Iwanaga
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| | - Nagisa Yahagi
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| | - Satoko Hakeda‐Suzuki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
- Research Initiatives and Promotion OrganizationYokohama National UniversityYokohamaJapan
| | - Takashi Suzuki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| |
Collapse
|
7
|
Kim N, Li Y, Yu R, Kwon HS, Song A, Jun MH, Jeong JY, Lee JH, Lim HH, Kim MJ, Kim JW, Oh WJ. Repulsive Sema3E-Plexin-D1 signaling coordinates both axonal extension and steering via activating an autoregulatory factor, Mtss1. eLife 2024; 13:e96891. [PMID: 38526535 PMCID: PMC11001299 DOI: 10.7554/elife.96891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
Axon guidance molecules are critical for neuronal pathfinding because they regulate directionality and growth pace during nervous system development. However, the molecular mechanisms coordinating proper axonal extension and turning are poorly understood. Here, metastasis suppressor 1 (Mtss1), a membrane protrusion protein, ensured axonal extension while sensitizing axons to the Semaphorin 3E (Sema3E)-Plexin-D1 repulsive cue. Sema3E-Plexin-D1 signaling enhanced Mtss1 expression in projecting striatonigral neurons. Mtss1 localized to the neurite axonal side and regulated neurite outgrowth in cultured neurons. Mtss1 also aided Plexin-D1 trafficking to the growth cone, where it signaled a repulsive cue to Sema3E. Mtss1 ablation reduced neurite extension and growth cone collapse in cultured neurons. Mtss1-knockout mice exhibited fewer striatonigral projections and irregular axonal routes, and these defects were recapitulated in Plxnd1- or Sema3e-knockout mice. These findings demonstrate that repulsive axon guidance activates an exquisite autoregulatory program coordinating both axonal extension and steering during neuronal pathfinding.
Collapse
Affiliation(s)
- Namsuk Kim
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Yan Li
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Ri Yu
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Hyo-Shin Kwon
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Anji Song
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Mi-Hee Jun
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Jin-Young Jeong
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and TechnologyDaeguRepublic of Korea
| | - Ji Hyun Lee
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Mi-Jin Kim
- Department of Life Sciences, Chung-Ang UniversitySeoulRepublic of Korea
| | - Jung-Woong Kim
- Department of Life Sciences, Chung-Ang UniversitySeoulRepublic of Korea
| | - Won-Jong Oh
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| |
Collapse
|
8
|
Nakajima C, Sawada M, Umeda E, Takagi Y, Nakashima N, Kuboyama K, Kaneko N, Yamamoto S, Nakamura H, Shimada N, Nakamura K, Matsuno K, Uesugi S, Vepřek NA, Küllmer F, Nasufović V, Uchiyama H, Nakada M, Otsuka Y, Ito Y, Herranz-Pérez V, García-Verdugo JM, Ohno N, Arndt HD, Trauner D, Tabata Y, Igarashi M, Sawamoto K. Identification of the growth cone as a probe and driver of neuronal migration in the injured brain. Nat Commun 2024; 15:1877. [PMID: 38461182 PMCID: PMC10924819 DOI: 10.1038/s41467-024-45825-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/01/2024] [Indexed: 03/11/2024] Open
Abstract
Axonal growth cones mediate axonal guidance and growth regulation. We show that migrating neurons in mice possess a growth cone at the tip of their leading process, similar to that of axons, in terms of the cytoskeletal dynamics and functional responsivity through protein tyrosine phosphatase receptor type sigma (PTPσ). Migrating-neuron growth cones respond to chondroitin sulfate (CS) through PTPσ and collapse, which leads to inhibition of neuronal migration. In the presence of CS, the growth cones can revert to their extended morphology when their leading filopodia interact with heparan sulfate (HS), thus re-enabling neuronal migration. Implantation of an HS-containing biomaterial in the CS-rich injured cortex promotes the extension of the growth cone and improve the migration and regeneration of neurons, thereby enabling functional recovery. Thus, the growth cone of migrating neurons is responsive to extracellular environments and acts as a primary regulator of neuronal migration.
Collapse
Affiliation(s)
- Chikako Nakajima
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Erika Umeda
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Yuma Takagi
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Norihiko Nakashima
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Kazuya Kuboyama
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Naoko Kaneko
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
- Laboratory of Neuronal Regeneration, Graduate School of Brain Science, Doshisha University, Kyoto, 610-0394, Japan
| | - Satoaki Yamamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Haruno Nakamura
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Naoki Shimada
- Research and Development Center, The Japan Wool Textile Co., Ltd., Kobe, 675-0053, Japan
| | - Koichiro Nakamura
- Medical Device Department, Nikke Medical Co., Ltd., Osaka, 541-0048, Japan
| | - Kumiko Matsuno
- Research and Development Center, The Japan Wool Textile Co., Ltd., Kobe, 675-0053, Japan
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences (LiMe), Kyoto University, Kyoto, 606-8507, Japan
| | - Shoji Uesugi
- Medical Device Department, Nikke Medical Co., Ltd., Osaka, 541-0048, Japan
| | - Nynke A Vepřek
- Department of Chemistry, New York University, New York, NY, 10003, USA
| | - Florian Küllmer
- Institute for Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University Jena, Jena, 07743, Germany
| | - Veselin Nasufović
- Institute for Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University Jena, Jena, 07743, Germany
| | | | | | - Yuji Otsuka
- Toray Research Center, Inc., Otsu, 520-8567, Japan
| | - Yasuyuki Ito
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Cavanilles Institute, University of Valencia, CIBERNED, Valencia, 46980, Spain
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute, University of Valencia, CIBERNED, Valencia, 46980, Spain
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, School of Medicine, Shimotsuke, 329-0498, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Hans-Dieter Arndt
- Institute for Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University Jena, Jena, 07743, Germany
| | - Dirk Trauner
- Department of Chemistry, New York University, New York, NY, 10003, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences (LiMe), Kyoto University, Kyoto, 606-8507, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan.
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan.
| |
Collapse
|
9
|
Tsai YC, Huang SM, Peng HH, Lin SW, Lin SR, Chin TY, Huang SM. Imbalance of synaptic and extrasynaptic NMDA receptors induced by the deletion of CRMP1 accelerates age-related cognitive decline in mice. Neurobiol Aging 2024; 135:48-59. [PMID: 38176125 DOI: 10.1016/j.neurobiolaging.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
Collapsin response mediator protein 1 (CRMP1) is involved in semaphorin 3A signaling pathway, promoting neurite extension and growth cone collapse. It is highly expressed in the nervous system, especially the hippocampus. The crmp1 knockout (KO) mice display impaired spatial learning and memory, and this phenomenon seemingly tends to deteriorate with age. Here we investigated whether CRMP1 is involved in age-related cognitive decline in WT and crmp1 KO mice at adult, middle-aged and older stages. The results revealed that cognitive dysfunction in the Morris water maze task became more severe and decreased glutamate and glutamine level in middle-aged crmp1 KO mice. Additionally, increasing levels of extrasynaptic NMDA receptors and phosphorylation of Tau were observed in middle-aged crmp1 KO mice, leading to synaptic and neuronal loss in the CA3 regions of hippocampus. These findings suggest that deletion of CRMP1 accelerates age-related cognitive decline by disrupting the balance between synaptic and extrasynaptic NMDA receptors, resulting in the loss of synapses and neurons.
Collapse
Affiliation(s)
- Yun-Chieh Tsai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Sheng-Min Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Hsu-Hsia Peng
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Rung Lin
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan.
| | - Ting-Yu Chin
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan.
| | - Shih-Ming Huang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan; Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
10
|
Qiu Z, Minegishi T, Aoki D, Abe K, Baba K, Inagaki N. Adhesion-clutch between DCC and netrin-1 mediates netrin-1-induced axonal haptotaxis. Front Mol Neurosci 2024; 17:1307755. [PMID: 38375502 PMCID: PMC10875621 DOI: 10.3389/fnmol.2024.1307755] [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: 10/05/2023] [Accepted: 01/08/2024] [Indexed: 02/21/2024] Open
Abstract
The growth cone, a motile structure located at the tip of growing axons, senses extracellular guidance cues and translates them into directional forces that drive axon outgrowth and guidance. Axon guidance directed by chemical cues on the extracellular adhesive substrate is termed haptotaxis. Recent studies reported that netrin-1 on the substrate functions as a haptotactic axon guidance cue. However, the mechanism mediating netrin-1-induced axonal haptotaxis remains unclear. Here, we demonstrate that substrate-bound netrin-1 induces axonal haptotaxis by facilitating physical interactions between the netrin-1 receptor, DCC, and the adhesive substrates. DCC serves as an adhesion receptor for netrin-1. The clutch-linker molecule shootin1a interacted with DCC, linking it to actin filament retrograde flow at the growth cone. Speckle imaging analyses showed that DCC underwent either grip (stop) or retrograde slip on the adhesive substrate. The grip state was more prevalent on netrin-1-coated substrate compared to the control substrate polylysine, thereby transmitting larger traction force on the netrin-1-coated substrate. Furthermore, disruption of the linkage between actin filament retrograde flow and DCC by shootin1 knockout impaired netrin-1-induced axonal haptotaxis. These results suggest that the directional force for netrin-1-induced haptotaxis is exerted on the substrates through the adhesion-clutch between DCC and netrin-1 which occurs asymmetrically within the growth cone.
Collapse
Affiliation(s)
| | | | | | | | | | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Nara, Japan
| |
Collapse
|
11
|
Yamamoto O, Saito R, Ohseki Y, Hoshino A. Nerve Regeneration and Gait Function Recovery with Implantation of Glucose/Mannose Conduits Using a Rat Model: Efficacy of Glucose/Mannose as a New Neurological Guidance Material. Bioengineering (Basel) 2024; 11:157. [PMID: 38391643 PMCID: PMC10886352 DOI: 10.3390/bioengineering11020157] [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: 12/27/2023] [Revised: 01/24/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Therapy with clinical nerve guidance conduits often causes functional incompleteness in patients. With the aim of better therapeutic efficacy, nerve regeneration and gait function were investigated in this study using a novel nerve guidance conduit consisting of glucose/mannose. The glucose/mannose nerve guidance conduits were prepared by filling the conduits with the glucose/mannose aqueous solutions for different kinematic viscosity, which were applied to sciatic nerve defects (6 mm gap) in a rat model. The nerve regeneration effect and the gait function recovery with the fabricated nerve guidance conduits were examined. From the results of the XRD measurement, the glucose/mannose conduits were identified as crystal structures of cellulose type II. Young's modulus and the maximum tensile strength of the crystalline glucose/mannose conduits demonstrated good strength and softness for the human nerve. Above 4 weeks postoperative, macroscopic observation revealed that the nerve was regenerated in the defective area. In various staining results of the nerve tissue removed at 4 weeks postoperative, myelinated nerves contributing to gait function could not be observed in the proximal and distal sites to the central nerve. At 8-12 weeks postoperative, myelinated nerves were found at the proximal and distal sites in hematoxylin/eosin staining. Glia cells were confirmed by phosphotungstic acid-hematoxylin staining. Continuous nerve fibers were observed clearly in the sections of the regenerated nerves towards the longitudinal direction at 12 weeks postoperative. The angle between the metatarsophalangeal joint and the ground plane was approximately 93° in intact rats. At 4 weeks postoperative, walking was not possible, but at 8 weeks postoperative, the rats were able to walk, with an angle of 53°. At 12 weeks postoperative, the angle increased further, reaching 65°, confirming that the rats were able to walk more quickly than at 8 weeks postoperative. These results demonstrated that gait function in rats treated with glucose/mannose nerve guidance conduits was rapidly recovered after 8 weeks postoperative. The glucose/mannose nerve guidance conduit could be applied as a new promising candidate material for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Osamu Yamamoto
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa 992-8510, Yamagata, Japan
| | - Risa Saito
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa 992-8510, Yamagata, Japan
| | - Yuta Ohseki
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa 992-8510, Yamagata, Japan
| | - Asami Hoshino
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa 992-8510, Yamagata, Japan
| |
Collapse
|
12
|
Niemsiri V, Rosenthal SB, Nievergelt CM, Maihofer AX, Marchetto MC, Santos R, Shekhtman T, Alliey-Rodriguez N, Anand A, Balaraman Y, Berrettini WH, Bertram H, Burdick KE, Calabrese JR, Calkin CV, Conroy C, Coryell WH, DeModena A, Eyler LT, Feeder S, Fisher C, Frazier N, Frye MA, Gao K, Garnham J, Gershon ES, Goes FS, Goto T, Harrington GJ, Jakobsen P, Kamali M, Kelly M, Leckband SG, Lohoff FW, McCarthy MJ, McInnis MG, Craig D, Millett CE, Mondimore F, Morken G, Nurnberger JI, Donovan CO, Øedegaard KJ, Ryan K, Schinagle M, Shilling PD, Slaney C, Stapp EK, Stautland A, Tarwater B, Zandi PP, Alda M, Fisch KM, Gage FH, Kelsoe JR. Focal adhesion is associated with lithium response in bipolar disorder: evidence from a network-based multi-omics analysis. Mol Psychiatry 2024; 29:6-19. [PMID: 36991131 PMCID: PMC11078741 DOI: 10.1038/s41380-022-01909-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 11/14/2022] [Accepted: 12/02/2022] [Indexed: 03/31/2023]
Abstract
Lithium (Li) is one of the most effective drugs for treating bipolar disorder (BD), however, there is presently no way to predict response to guide treatment. The aim of this study is to identify functional genes and pathways that distinguish BD Li responders (LR) from BD Li non-responders (NR). An initial Pharmacogenomics of Bipolar Disorder study (PGBD) GWAS of lithium response did not provide any significant results. As a result, we then employed network-based integrative analysis of transcriptomic and genomic data. In transcriptomic study of iPSC-derived neurons, 41 significantly differentially expressed (DE) genes were identified in LR vs NR regardless of lithium exposure. In the PGBD, post-GWAS gene prioritization using the GWA-boosting (GWAB) approach identified 1119 candidate genes. Following DE-derived network propagation, there was a highly significant overlap of genes between the top 500- and top 2000-proximal gene networks and the GWAB gene list (Phypergeometric = 1.28E-09 and 4.10E-18, respectively). Functional enrichment analyses of the top 500 proximal network genes identified focal adhesion and the extracellular matrix (ECM) as the most significant functions. Our findings suggest that the difference between LR and NR was a much greater effect than that of lithium. The direct impact of dysregulation of focal adhesion on axon guidance and neuronal circuits could underpin mechanisms of response to lithium, as well as underlying BD. It also highlights the power of integrative multi-omics analysis of transcriptomic and genomic profiling to gain molecular insights into lithium response in BD.
Collapse
Grants
- R01 MH095741 NIMH NIH HHS
- UL1 TR001442 NCATS NIH HHS
- I01 BX003431 BLRD VA
- U19 MH106434 NIMH NIH HHS
- U01 MH092758 NIMH NIH HHS
- T32 MH018399 NIMH NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- Department of Veterans Affairs | Veterans Affairs San Diego Healthcare System (VA San Diego Healthcare System)
- The Halifax group (MA, CVC, JG, CO, and CS) is supported by grants from Canadian Institutes of Health Research (#166098), ERA PerMed project PLOT-BD, Research Nova Scotia, Genome Atlantic, Nova Scotia Health Authority and Dalhousie Medical Research Foundation (Lindsay Family Fund).
- U.S. Department of Health & Human Services | NIH | National Center for Advancing Translational Sciences (NCATS)
- U19MH106434, part of the National Cooperative Reprogrammed Cell Research Groups (NCRCRG) to Study Mental Illness. AHA-Allen Initiative in Brain Health and Cognitive Impairment Award (19PABH134610000). The JPB Foundation, Bob and Mary Jane Engman, Annette C Merle-Smith, R01 MH095741, and Lynn and Edward Streim.
Collapse
Affiliation(s)
- Vipavee Niemsiri
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Sara Brin Rosenthal
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | | | - Adam X Maihofer
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Maria C Marchetto
- Department of Anthropology, University of California, San Diego, La Jolla, CA, USA
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Renata Santos
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
- University of Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1261266, Laboratory of Dynamics of Neuronal Structure in Health and Disease, Paris, France
| | - Tatyana Shekhtman
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Ney Alliey-Rodriguez
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
- Department of Psychiatry and Behavioral Neuroscience, Northwestern University, Chicago, IL, USA
| | - Amit Anand
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yokesh Balaraman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wade H Berrettini
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Holli Bertram
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Katherine E Burdick
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph R Calabrese
- Mood Disorders Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Mood Disorders Program, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Cynthia V Calkin
- Department of Psychiatry and Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Carla Conroy
- Mood Disorders Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Mood Disorders Program, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | - Anna DeModena
- Psychiatry Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Lisa T Eyler
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Scott Feeder
- Department of Psychiatry, The Mayo Clinic, Rochester, MN, USA
| | - Carrie Fisher
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicole Frazier
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Mark A Frye
- Department of Psychiatry, The Mayo Clinic, Rochester, MN, USA
| | - Keming Gao
- Mood Disorders Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Mood Disorders Program, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Julie Garnham
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Elliot S Gershon
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Toyomi Goto
- Mood Disorders Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Petter Jakobsen
- Norment, Division of Psychiatry, Haukeland University Hospital and Department of Clinical medicine, University of Bergen, Bergen, Norway
| | - Masoud Kamali
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Marisa Kelly
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Susan G Leckband
- Psychiatry Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Falk W Lohoff
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J McCarthy
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Melvin G McInnis
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - David Craig
- Department of Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - Caitlin E Millett
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francis Mondimore
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Gunnar Morken
- Division of Mental Health Care, St Olavs University Hospital, and Department of Mental Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - John I Nurnberger
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Ketil J Øedegaard
- Norment, Division of Psychiatry, Haukeland University Hospital and Department of Clinical medicine, University of Bergen, Bergen, Norway
| | - Kelly Ryan
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Martha Schinagle
- Mood Disorders Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Paul D Shilling
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Claire Slaney
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Emma K Stapp
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Andrea Stautland
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Bruce Tarwater
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Peter P Zandi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- National Institute of Mental Health, Klecany, Czech Republic
| | - Kathleen M Fisch
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - John R Kelsoe
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
13
|
Huang H, Majumder T, Khot B, Suriyaarachchi H, Yang T, Shao Q, Tirukovalluru S, Liu G. The role of microtubule-associated protein tau in netrin-1 attractive signaling. J Cell Sci 2024; 137:jcs261244. [PMID: 38197773 PMCID: PMC10906489 DOI: 10.1242/jcs.261244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/24/2023] [Indexed: 01/11/2024] Open
Abstract
Direct binding of netrin receptors with dynamic microtubules (MTs) in the neuronal growth cone plays an important role in netrin-mediated axon guidance. However, how netrin-1 (NTN1) regulates MT dynamics in axon turning remains a major unanswered question. Here, we show that the coupling of netrin-1 receptor DCC with tau (MAPT)-regulated MTs is involved in netrin-1-promoted axon attraction. Tau directly interacts with DCC and partially overlaps with DCC in the growth cone of primary neurons. Netrin-1 induces this interaction and the colocalization of DCC and tau in the growth cone. The netrin-1-induced interaction of tau with DCC relies on MT dynamics and TUBB3, a highly dynamic β-tubulin isotype in developing neurons. Netrin-1 increased cosedimentation of DCC with tau and TUBB3 in MTs, and knockdown of either tau or TUBB3 mutually blocked this effect. Downregulation of endogenous tau levels by tau shRNAs inhibited netrin-1-induced axon outgrowth, branching and commissural axon attraction in vitro, and led to defects in spinal commissural axon projection in vivo. These findings suggest that tau is a key MT-associated protein coupling DCC with MT dynamics in netrin-1-promoted axon attraction.
Collapse
Affiliation(s)
- Huai Huang
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Bhakti Khot
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Harindi Suriyaarachchi
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Shraddha Tirukovalluru
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| |
Collapse
|
14
|
Meza U, Romero-Méndez C, Sánchez-Armáss S, Rodríguez-Menchaca AA. Role of rafts in neurological disorders. Neurologia 2023; 38:671-680. [PMID: 37858892 DOI: 10.1016/j.nrleng.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/01/2021] [Indexed: 10/21/2023] Open
Abstract
INTRODUCTION Rafts are protein-lipid structural nanodomains involved in efficient signal transduction and the modulation of physiological processes of the cell plasma membrane. Raft disruption in the nervous system has been associated with a wide range of disorders. DEVELOPMENT We review the concept of rafts, the nervous system processes in which they are involved, and their role in diseases such as Parkinson's disease, Alzheimer disease, and Huntington disease. CONCLUSIONS Based on the available evidence, preservation and/or reconstitution of rafts is a promising treatment strategy for a wide range of neurological disorders.
Collapse
Affiliation(s)
- U Meza
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - C Romero-Méndez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - S Sánchez-Armáss
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - A A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| |
Collapse
|
15
|
Zhou FY, Weems A, Gihana GM, Chen B, Chang BJ, Driscoll M, Danuser G. Surface-guided computing to analyze subcellular morphology and membrane-associated signals in 3D. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536640. [PMID: 37131779 PMCID: PMC10153113 DOI: 10.1101/2023.04.12.536640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Signal transduction and cell function are governed by the spatiotemporal organization of membrane-associated molecules. Despite significant advances in visualizing molecular distributions by 3D light microscopy, cell biologists still have limited quantitative understanding of the processes implicated in the regulation of molecular signals at the whole cell scale. In particular, complex and transient cell surface morphologies challenge the complete sampling of cell geometry, membrane-associated molecular concentration and activity and the computing of meaningful parameters such as the cofluctuation between morphology and signals. Here, we introduce u-Unwrap3D, a framework to remap arbitrarily complex 3D cell surfaces and membrane-associated signals into equivalent lower dimensional representations. The mappings are bidirectional, allowing the application of image processing operations in the data representation best suited for the task and to subsequently present the results in any of the other representations, including the original 3D cell surface. Leveraging this surface-guided computing paradigm, we track segmented surface motifs in 2D to quantify the recruitment of Septin polymers by blebbing events; we quantify actin enrichment in peripheral ruffles; and we measure the speed of ruffle movement along topographically complex cell surfaces. Thus, u-Unwrap3D provides access to spatiotemporal analyses of cell biological parameters on unconstrained 3D surface geometries and signals.
Collapse
Affiliation(s)
- Felix Y. Zhou
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Weems
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabriel M. Gihana
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bingying Chen
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bo-Jui Chang
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Meghan Driscoll
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Current address: Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
16
|
Zhou FY, Weems A, Gihana GM, Chen B, Chang BJ, Driscoll M, Danuser G. Surface-guided computing to analyze subcellular morphology and membrane-associated signals in 3D. ARXIV 2023:arXiv:2304.06176v1. [PMID: 37090235 PMCID: PMC10120750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Signal transduction and cell function are governed by the spatiotemporal organization of membrane-associated molecules. Despite significant advances in visualizing molecular distributions by 3D light microscopy, cell biologists still have limited quantitative understanding of the processes implicated in the regulation of molecular signals at the whole cell scale. In particular, complex and transient cell surface morphologies challenge the complete sampling of cell geometry, membrane-associated molecular concentration and activity and the computing of meaningful parameters such as the cofluctuation between morphology and signals. Here, we introduce u-Unwrap3D, a framework to remap arbitrarily complex 3D cell surfaces and membrane-associated signals into equivalent lower dimensional representations. The mappings are bidirectional, allowing the application of image processing operations in the data representation best suited for the task and to subsequently present the results in any of the other representations, including the original 3D cell surface. Leveraging this surface-guided computing paradigm, we track segmented surface motifs in 2D to quantify the recruitment of Septin polymers by blebbing events; we quantify actin enrichment in peripheral ruffles; and we measure the speed of ruffle movement along topographically complex cell surfaces. Thus, u-Unwrap3D provides access to spatiotemporal analyses of cell biological parameters on unconstrained 3D surface geometries and signals.
Collapse
Affiliation(s)
- Felix Y. Zhou
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Weems
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabriel M. Gihana
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bingying Chen
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bo-Jui Chang
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Meghan Driscoll
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Current address: Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
17
|
Kastian RF, Baba K, Kaewkascholkul N, Sasaki H, Watanabe R, Toriyama M, Inagaki N. Dephosphorylation of neural wiring protein shootin1 by PP1 phosphatase regulates netrin-1-induced axon guidance. J Biol Chem 2023; 299:104687. [PMID: 37044214 DOI: 10.1016/j.jbc.2023.104687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023] Open
Abstract
Axon pathfinding is an essential step in neuronal network formation. Shootin1a is a clutch-linker molecule that is mechanically involved in axon outgrowth and guidance. It was previously shown that concentration gradients of axon guidance molecule netrin-1 in the extracellular environment elicit asymmetrically localized Pak1 kinase-mediated phosphorylation of shootin1a within axonal growth cones, which is higher on the netrin-1 source side. This asymmetric phosphorylation promotes shootin1a-mediated local actin-adhesion coupling within growth cones, thereby generating directional forces for turning the growth cone toward the netrin-1 source. However, how the spatial differences in netrin-1 concentration are transduced into the asymmetrically localized signaling within growth cones remains unclear. Moreover, the protein phosphatases that dephosphorylate shootin1a remain unidentified. Here, we report that protein phosphatase-1 (PP1) dephosphorylates shootin1a in growth cones. We found that PP1 overexpression abolished the netrin-1-induced asymmetric localization of phosphorylated-shootin1a as well as axon turning. In addition, we show PP1 inhibition reversed the asymmetrically localized shootin1a phosphorylation within growth cones under netrin-1 gradient, thereby changing the netrin-1-induced growth cone turning from attraction to repulsion. These data indicate that PP1-mediated shootin1a dephosphorylation plays a key role in organizing asymmetrically-localized phosphorylated shootin1a within growth cones, which regulates netrin-1-induced axon guidance.
Collapse
Affiliation(s)
- Ria Fajarwati Kastian
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan; Mammalian Cell Engineering and Signal Transduction Research Group, Research Center for Genetic Engineering, National Research and Innovation Agency, KST Soekarno, Jl. Raya Bogor, KM. 46, Cibinong, Bogor, West Java, 16911, Indonesia
| | - Kentarou Baba
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Napol Kaewkascholkul
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Hisashi Sasaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Rikiya Watanabe
- Molecular Physiology Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Michinori Toriyama
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan.
| |
Collapse
|
18
|
Dema A, Charafeddine R, Rahgozar S, van Haren J, Wittmann T. Growth cone advance requires EB1 as revealed by genomic replacement with a light-sensitive variant. eLife 2023; 12:84143. [PMID: 36715499 PMCID: PMC9917429 DOI: 10.7554/elife.84143] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
A challenge in analyzing dynamic intracellular cell biological processes is the dearth of methodologies that are sufficiently fast and specific to perturb intracellular protein activities. We previously developed a light-sensitive variant of the microtubule plus end-tracking protein EB1 by inserting a blue light-controlled protein dimerization module between functional domains. Here, we describe an advanced method to replace endogenous EB1 with this light-sensitive variant in a single genome editing step, thereby enabling this approach in human induced pluripotent stem cells (hiPSCs) and hiPSC-derived neurons. We demonstrate that acute and local optogenetic EB1 inactivation in developing cortical neurons induces microtubule depolymerization in the growth cone periphery and subsequent neurite retraction. In addition, advancing growth cones are repelled from areas of blue light exposure. These phenotypes were independent of the neuronal EB1 homolog EB3, revealing a direct dynamic role of EB1-mediated microtubule plus end interactions in neuron morphogenesis and neurite guidance.
Collapse
Affiliation(s)
- Alessandro Dema
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | - Rabab Charafeddine
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | - Shima Rahgozar
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | | | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
19
|
Schneider F, Metz I, Rust MB. Regulation of actin filament assembly and disassembly in growth cone motility and axon guidance. Brain Res Bull 2023; 192:21-35. [PMID: 36336143 DOI: 10.1016/j.brainresbull.2022.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Directed outgrowth of axons is fundamental for the establishment of neuronal networks. Axon outgrowth is guided by growth cones, highly motile structures enriched in filamentous actin (F-actin) located at the axons' distal tips. Growth cones exploit F-actin-based protrusions to scan the environment for guidance cues, and they contain the sensory apparatus to translate guidance cue information into intracellular signaling cascades. These cascades act upstream of actin-binding proteins (ABP) and thereby control assembly and disassembly of F-actin. Spatiotemporally controlled F-actin dis-/assembly in growth cones steers the axon towards attractants and away from repellents, and it thereby navigates the axon through the developing nervous system. Hence, ABP that control F-actin dynamics emerged as critical regulators of neuronal network formation. In the present review article, we will summarize and discuss current knowledge of the mechanisms that control remodeling of the actin cytoskeleton in growth cones, focusing on recent progress in the field. Further, we will introduce tools and techniques that allow to study actin regulatory mechanism in growth cones.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Molecular Urooncology, Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany.
| |
Collapse
|
20
|
Liaci C, Camera M, Zamboni V, Sarò G, Ammoni A, Parmigiani E, Ponzoni L, Hidisoglu E, Chiantia G, Marcantoni A, Giustetto M, Tomagra G, Carabelli V, Torelli F, Sala M, Yanagawa Y, Obata K, Hirsch E, Merlo GR. Loss of ARHGAP15 affects the directional control of migrating interneurons in the embryonic cortex and increases susceptibility to epilepsy. Front Cell Dev Biol 2022; 10:875468. [PMID: 36568982 PMCID: PMC9774038 DOI: 10.3389/fcell.2022.875468] [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: 02/14/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022] Open
Abstract
GTPases of the Rho family are components of signaling pathways linking extracellular signals to the control of cytoskeleton dynamics. Among these, RAC1 plays key roles during brain development, ranging from neuronal migration to neuritogenesis, synaptogenesis, and plasticity. RAC1 activity is positively and negatively controlled by guanine nucleotide exchange factors (GEFs), guanosine nucleotide dissociation inhibitors (GDIs), and GTPase-activating proteins (GAPs), but the specific role of each regulator in vivo is poorly known. ARHGAP15 is a RAC1-specific GAP expressed during development in a fraction of migrating cortical interneurons (CINs) and in the majority of adult CINs. During development, loss of ARHGAP15 causes altered directionality of the leading process of tangentially migrating CINs, along with altered morphology in vitro. Likewise, time-lapse imaging of embryonic CINs revealed a poorly coordinated directional control during radial migration, possibly due to a hyper-exploratory behavior. In the adult cortex, the observed defects lead to subtle alteration in the distribution of CALB2-, SST-, and VIP-positive interneurons. Adult Arhgap15-knock-out mice also show reduced CINs intrinsic excitability, spontaneous subclinical seizures, and increased susceptibility to the pro-epileptic drug pilocarpine. These results indicate that ARHGAP15 imposes a fine negative regulation on RAC1 that is required for morphological maturation and directional control during CIN migration, with consequences on their laminar distribution and inhibitory function.
Collapse
Affiliation(s)
- Carla Liaci
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Mattia Camera
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Valentina Zamboni
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Gabriella Sarò
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Alessandra Ammoni
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | | | - Luisa Ponzoni
- Neuroscience Institute, Consiglio Nazionale Ricerche, Milan, Italy
| | - Enis Hidisoglu
- Department of Drug Science, NIS Center, University of Turin, Turin, Italy
| | - Giuseppe Chiantia
- Department of Neuroscience and National Institute of Neuroscience, University of Turin, Turin, Italy
| | - Andrea Marcantoni
- Department of Drug Science, NIS Center, University of Turin, Turin, Italy
| | - Maurizio Giustetto
- Department of Neuroscience and National Institute of Neuroscience, University of Turin, Turin, Italy
| | - Giulia Tomagra
- Department of Drug Science, NIS Center, University of Turin, Turin, Italy
| | | | - Federico Torelli
- Institute for Physiology I, Medical Faculty, Albert-Ludwigs-University Freiburg, Freiburg, Germany,Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Mariaelvina Sala
- Neuroscience Institute, Consiglio Nazionale Ricerche, Milan, Italy
| | - Yuchio Yanagawa
- Department of Genetic Behavioral Neuroscience, Gunma University, Maebashi, Japan
| | | | - Emilio Hirsch
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Giorgio R. Merlo
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy,*Correspondence: Giorgio R. Merlo,
| |
Collapse
|
21
|
Li X, Shim S, Hardin KR, Vanaja KG, Song H, Levchenko A, Ming GL, Zheng JQ. Signal amplification in growth cone gradient sensing by a double negative feedback loop among PTEN, PI(3,4,5)P 3 and actomyosin. Mol Cell Neurosci 2022; 123:103772. [PMID: 36055521 PMCID: PMC9856701 DOI: 10.1016/j.mcn.2022.103772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 01/24/2023] Open
Abstract
Axon guidance during neural wiring involves a series of precisely controlled chemotactic events by the motile axonal tip, the growth cone. A fundamental question is how neuronal growth cones make directional decisions in response to extremely shallow gradients of guidance cues with exquisite sensitivity. Here we report that nerve growth cones possess a signal amplification mechanism during gradient sensing process. In neuronal growth cones of Xenopus spinal neurons, phosphatidylinositol-3,4,5-trisphosphate (PIP3), an important signaling molecule in chemotaxis, was actively recruited to the up-gradient side in response to an external gradient of brain-derived neurotrophic factor (BDNF), resulting in an intracellular gradient with approximate 30-fold amplification of the input. Furthermore, a reverse gradient of phosphatase and tensin homolog (PTEN) was induced by BDNF within the growth cone and the increased PTEN activity at the down-gradient side is required for the amplification of PIP3 signals. Mechanistically, the establishment of both positive PIP3 and reverse PTEN gradients depends on the filamentous actin network. Together with computational modeling, our results revealed a double negative feedback loop among PTEN, PIP3 and actomyosin for signal amplification, which is essential for gradient sensing of neuronal growth cones in response to diffusible cues.
Collapse
Affiliation(s)
- Xiong Li
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Sangwoo Shim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA; Department of Cell Biology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Katherine R Hardin
- Department of Cell Biology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Kiran G Vanaja
- Department of Biomedical Engineering and Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Andre Levchenko
- Department of Biomedical Engineering and Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - James Q Zheng
- Department of Cell Biology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA.
| |
Collapse
|
22
|
Triantopoulou N, Vidaki M. Local mRNA translation and cytoskeletal reorganization: Mechanisms that tune neuronal responses. Front Mol Neurosci 2022; 15:949096. [PMID: 35979146 PMCID: PMC9376447 DOI: 10.3389/fnmol.2022.949096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/07/2022] [Indexed: 12/31/2022] Open
Abstract
Neurons are highly polarized cells with significantly long axonal and dendritic extensions that can reach distances up to hundreds of centimeters away from the cell bodies in higher vertebrates. Their successful formation, maintenance, and proper function highly depend on the coordination of intricate molecular networks that allow axons and dendrites to quickly process information, and respond to a continuous and diverse cascade of environmental stimuli, often without enough time for communication with the soma. Two seemingly unrelated processes, essential for these rapid responses, and thus neuronal homeostasis and plasticity, are local mRNA translation and cytoskeletal reorganization. The axonal cytoskeleton is characterized by high stability and great plasticity; two contradictory attributes that emerge from the powerful cytoskeletal rearrangement dynamics. Cytoskeletal reorganization is crucial during nervous system development and in adulthood, ensuring the establishment of proper neuronal shape and polarity, as well as regulating intracellular transport and synaptic functions. Local mRNA translation is another mechanism with a well-established role in the developing and adult nervous system. It is pivotal for axonal guidance and arborization, synaptic formation, and function and seems to be a key player in processes activated after neuronal damage. Perturbations in the regulatory pathways of local translation and cytoskeletal reorganization contribute to various pathologies with diverse clinical manifestations, ranging from intellectual disabilities (ID) to autism spectrum disorders (ASD) and schizophrenia (SCZ). Despite the fact that both processes are essential for the orchestration of pathways critical for proper axonal and dendritic function, the interplay between them remains elusive. Here we review our current knowledge on the molecular mechanisms and specific interaction networks that regulate and potentially coordinate these interconnected processes.
Collapse
Affiliation(s)
- Nikoletta Triantopoulou
- Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
| | - Marina Vidaki
- Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
- *Correspondence: Marina Vidaki,
| |
Collapse
|
23
|
Hylton RK, Heebner JE, Grillo MA, Swulius MT. Cofilactin filaments regulate filopodial structure and dynamics in neuronal growth cones. Nat Commun 2022; 13:2439. [PMID: 35508487 PMCID: PMC9068697 DOI: 10.1038/s41467-022-30116-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 04/15/2022] [Indexed: 12/12/2022] Open
Abstract
Cofilin is best known for its ability to sever actin filaments and facilitate cytoskeletal recycling inside of cells, but at higher concentrations in vitro, cofilin stabilizes a more flexible, hyper-twisted state of actin known as “cofilactin”. While this filament state is well studied, a structural role for cofilactin in dynamic cellular processes has not been observed. With a combination of cryo-electron tomography and fluorescence imaging in neuronal growth cones, we observe that filopodial actin filaments switch between a fascin-linked and a cofilin-decorated state, and that cofilactin is associated with a variety of dynamic events within filopodia. The switch to cofilactin filaments occurs in a graded fashion and correlates with a decline in fascin cross-linking within the filopodia, which is associated with curvature in the bundle. Our tomographic data reveal that the hyper-twisting of actin from cofilin binding leads to a rearrangement of filament packing, which largely excludes fascin from the base of filopodia. Our results provide mechanistic insight into the fundamentals of cytoskeletal remodeling inside of confined cellular spaces, and how the interplay between fascin and cofilin regulates the dynamics of searching filopodia. In this manuscript the authors show that Filopodia switch between bundles of fascin-crosslinked actin and cofilin-decorated filaments, which exclude fascin binding due to altered structure and packing, as well as affect filopodial searching dynamics.
Collapse
Affiliation(s)
- Ryan K Hylton
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
| | - Jessica E Heebner
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
| | - Michael A Grillo
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
| | - Matthew T Swulius
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
24
|
Gatford NJF, Deans PJM, Duarte RRR, Chennell G, Sellers KJ, Raval P, Srivastava DP. Neuroligin-3 and neuroligin-4X form nanoscopic clusters and regulate growth cone organization and size. Hum Mol Genet 2022; 31:674-691. [PMID: 34542148 PMCID: PMC8895740 DOI: 10.1093/hmg/ddab277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/25/2021] [Accepted: 09/13/2021] [Indexed: 12/01/2022] Open
Abstract
The cell-adhesion proteins neuroligin-3 and neuroligin-4X (NLGN3/4X) have well described roles in synapse formation. NLGN3/4X are also expressed highly during neurodevelopment. However, the role these proteins play during this period is unknown. Here we show that NLGN3/4X localized to the leading edge of growth cones where it promoted neuritogenesis in immature human neurons. Super-resolution microscopy revealed that NLGN3/4X clustering induced growth cone enlargement and influenced actin filament organization. Critically, these morphological effects were not induced by autism spectrum disorder (ASD)-associated NLGN3/4X variants. Finally, actin regulators p21-activated kinase 1 and cofilin were found to be activated by NLGN3/4X and involved in mediating the effects of these adhesion proteins on actin filaments, growth cones and neuritogenesis. These data reveal a novel role for NLGN3 and NLGN4X in the development of neuronal architecture, which may be altered in the presence of ASD-associated variants.
Collapse
Affiliation(s)
- Nicholas J F Gatford
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - P J Michael Deans
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Rodrigo R R Duarte
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| | - George Chennell
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| | - Katherine J Sellers
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
25
|
Engmann AK, Hatch JJ, Nanda P, Veeraraghavan P, Ozkan A, Poulopoulos A, Murphy AJ, Macklis JD. Neuronal subtype-specific growth cone and soma purification from mammalian CNS via fractionation and fluorescent sorting for subcellular analyses and spatial mapping of local transcriptomes and proteomes. Nat Protoc 2022; 17:222-251. [PMID: 35022617 PMCID: PMC9751848 DOI: 10.1038/s41596-021-00638-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 09/24/2021] [Indexed: 11/09/2022]
Abstract
During neuronal development, growth cones (GCs) of projection neurons navigate complex extracellular environments to reach distant targets, thereby generating extraordinarily complex circuitry. These dynamic structures located at the tips of axonal projections respond to substrate-bound as well as diffusible guidance cues in a neuronal subtype- and stage-specific manner to construct highly specific and functional circuitry. In vitro studies of the past decade indicate that subcellular localization of specific molecular machinery in GCs underlies the precise navigational control that occurs during circuit 'wiring'. Our laboratory has recently developed integrated experimental and analytical approaches enabling high-depth, quantitative proteomic and transcriptomic investigation of subtype- and stage-specific GC molecular machinery directly from the rodent central nervous system (CNS) in vivo. By using these approaches, a pure population of GCs and paired somata can be isolated from any neuronal subtype of the CNS that can be fluorescently labeled. GCs are dissociated from parent axons using fluid shear forces, and a bulk GC fraction is isolated by buoyancy ultracentrifugation. Subtype-specific GCs and somata are purified by recently developed fluorescent small particle sorting and established FACS of neurons and are suitable for downstream analyses of proteins and RNAs, including small RNAs. The isolation of subtype-specific GCs and parent somata takes ~3 h, plus sorting time, and ~1-2 h for subsequent extraction of molecular contents. RNA library preparation and sequencing can take several days to weeks, depending on the turnaround time of the core facility involved.
Collapse
Affiliation(s)
- Anne K Engmann
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - John J Hatch
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Prakruti Nanda
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Priya Veeraraghavan
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Abdulkadir Ozkan
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Alexandros Poulopoulos
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alexander J Murphy
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
- Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
26
|
Sánchez-Huertas C, Herrera E. With the Permission of Microtubules: An Updated Overview on Microtubule Function During Axon Pathfinding. Front Mol Neurosci 2021; 14:759404. [PMID: 34924953 PMCID: PMC8675249 DOI: 10.3389/fnmol.2021.759404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/27/2023] Open
Abstract
During the establishment of neural circuitry axons often need to cover long distances to reach remote targets. The stereotyped navigation of these axons defines the connectivity between brain regions and cellular subtypes. This chemotrophic guidance process mostly relies on the spatio-temporal expression patterns of extracellular proteins and the selective expression of their receptors in projection neurons. Axon guidance is stimulated by guidance proteins and implemented by neuronal traction forces at the growth cones, which engage local cytoskeleton regulators and cell adhesion proteins. Different layers of guidance signaling regulation, such as the cleavage and processing of receptors, the expression of co-receptors and a wide variety of intracellular cascades downstream of receptors activation, have been progressively unveiled. Also, in the last decades, the regulation of microtubule (MT) assembly, stability and interactions with the submembranous actin network in the growth cone have emerged as crucial effector mechanisms in axon pathfinding. In this review, we will delve into the intracellular signaling cascades downstream of guidance receptors that converge on the MT cytoskeleton of the growing axon. In particular, we will focus on the microtubule-associated proteins (MAPs) network responsible of MT dynamics in the axon and growth cone. Complementarily, we will discuss new evidences that connect defects in MT scaffold proteins, MAPs or MT-based motors and axon misrouting during brain development.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Alicante, Spain
| | | |
Collapse
|
27
|
Brown SL, Ren Y, Suter DM, Mattoo S. A Co-purification Method for Efficient Production and Src Kinase-mediated Phosphorylation of Aplysia Cortactin. Bio Protoc 2021; 11:e4158. [PMID: 34692908 PMCID: PMC8481015 DOI: 10.21769/bioprotoc.4158] [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: 03/05/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 11/02/2022] Open
Abstract
Cortactin is an actin-binding protein that regulates processes like cell migration, endocytosis, and tumor cell metastasis. Although cortactin is associated with actin-cytoskeletal dynamics in non-neuronal cells and cell-free systems, the exact mechanisms underlying its fundamental roles in neuronal growth cones are not fully explored. Recent reports show that Aplysia Src2 tyrosine kinase induces phosphorylation of cortactin as a mechanism to control lamellipodia protrusion and filopodia formation in cultured Aplysia bag cell neurons ( He et al., 2015 ; Ren et al., 2019 ). In order to provide in vitro evidence for Src2-mediated phosphorylation of cortactin, we developed a robust and cost-effective method for the efficient expression and purification of Aplysia cortactin and Src2 kinase that can be used for biochemical studies including phosphorylation assays. By co-purifying cortactin and Src kinase with a phosphatase (YopH) from Yersinia enterocolitica, we eliminated the problem of non-specific phosphorylation of induced proteins by bacterial kinases and also reduced costs by bypassing the need for commercial enzymatic treatments. This protocol is reproducible and can be modified to produce homogenous non-phosphorylated proteins during recombinant protein expression in Escherichia coli.
Collapse
Affiliation(s)
- Sherlene L Brown
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Yuan Ren
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA.,Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.,Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| | - Seema Mattoo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA.,Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
28
|
Agrawal M, Welshhans K. Local Translation Across Neural Development: A Focus on Radial Glial Cells, Axons, and Synaptogenesis. Front Mol Neurosci 2021; 14:717170. [PMID: 34434089 PMCID: PMC8380849 DOI: 10.3389/fnmol.2021.717170] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
In the past two decades, significant progress has been made in our understanding of mRNA localization and translation at distal sites in axons and dendrites. The existing literature shows that local translation is regulated in a temporally and spatially restricted manner and is critical throughout embryonic and post-embryonic life. Here, recent key findings about mRNA localization and local translation across the various stages of neural development, including neurogenesis, axon development, and synaptogenesis, are reviewed. In the early stages of development, mRNAs are localized and locally translated in the endfeet of radial glial cells, but much is still unexplored about their functional significance. Recent in vitro and in vivo studies have provided new information about the specific mechanisms regulating local translation during axon development, including growth cone guidance and axon branching. Later in development, localization and translation of mRNAs help mediate the major structural and functional changes that occur in the axon during synaptogenesis. Clinically, changes in local translation across all stages of neural development have important implications for understanding the etiology of several neurological disorders. Herein, local translation and mechanisms regulating this process across developmental stages are compared and discussed in the context of function and dysfunction.
Collapse
Affiliation(s)
- Manasi Agrawal
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Kristy Welshhans
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
29
|
Unraveling Axon Guidance during Axotomy and Regeneration. Int J Mol Sci 2021; 22:ijms22158344. [PMID: 34361110 PMCID: PMC8347220 DOI: 10.3390/ijms22158344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
During neuronal development and regeneration axons extend a cytoskeletal-rich structure known as the growth cone, which detects and integrates signals to reach its final destination. The guidance cues “signals” bind their receptors, activating signaling cascades that result in the regulation of the growth cone cytoskeleton, defining growth cone advance, pausing, turning, or collapse. Even though much is known about guidance cues and their isolated mechanisms during nervous system development, there is still a gap in the understanding of the crosstalk between them, and about what happens after nervous system injuries. After neuronal injuries in mammals, only axons in the peripheral nervous system are able to regenerate, while the ones from the central nervous system fail to do so. Therefore, untangling the guidance cues mechanisms, as well as their behavior and characterization after axotomy and regeneration, are of special interest for understanding and treating neuronal injuries. In this review, we present findings on growth cone guidance and canonical guidance cues mechanisms, followed by a description and comparison of growth cone pathfinding mechanisms after axotomy, in regenerative and non-regenerative animal models.
Collapse
|
30
|
Dumoulin A, Zuñiga NR, Stoeckli ET. Axon guidance at the spinal cord midline-A live imaging perspective. J Comp Neurol 2021; 529:2517-2538. [PMID: 33438755 PMCID: PMC8248161 DOI: 10.1002/cne.25107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/10/2020] [Accepted: 01/08/2021] [Indexed: 12/19/2022]
Abstract
During neural circuit formation, axons navigate several choice points to reach their final target. At each one of these intermediate targets, growth cones need to switch responsiveness from attraction to repulsion in order to move on. Molecular mechanisms that allow for the precise timing of surface expression of a new set of receptors that support the switch in responsiveness are difficult to study in vivo. Mostly, mechanisms are inferred from the observation of snapshots of many different growth cones analyzed in different preparations of tissue harvested at distinct time points. However, to really understand the behavior of growth cones at choice points, a single growth cone should be followed arriving at and leaving the intermediate target. Existing ex vivo preparations, like cultures of an "open-book" preparation of the spinal cord have been successfully used to study floor plate entry and exit, but artifacts prevent the analysis of growth cone behavior at the floor plate exit site. Here, we describe a novel spinal cord preparation that allows for live imaging of individual axons during navigation in their intact environment. When comparing growth cone behavior in our ex vivo system with snapshots from in vivo navigation, we do not see any differences. The possibility to observe the dynamics of single growth cones navigating their intermediate target allows for measuring growth speed, changes in morphology, or aberrant behavior, like stalling and wrong turning. Moreover, observation of the intermediate target-the floor plate-revealed its active participation and interaction with commissural axons during midline crossing.
Collapse
Affiliation(s)
- Alexandre Dumoulin
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of ZurichZurichSwitzerland
| | - Nikole R. Zuñiga
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of ZurichZurichSwitzerland
| | - Esther T. Stoeckli
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of ZurichZurichSwitzerland
| |
Collapse
|
31
|
Kundu T, Dutta P, Nagar D, Maiti S, Ghose A. Coupling of dynamic microtubules to F-actin by Fmn2 regulates chemotaxis of neuronal growth cones. J Cell Sci 2021; 134:jcs252916. [PMID: 34313311 DOI: 10.1242/jcs.252916] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Dynamic co-regulation of the actin and microtubule subsystems enables the highly precise and adaptive remodelling of the cytoskeleton necessary for critical cellular processes, such as axonal pathfinding. The modes and mediators of this interpolymer crosstalk, however, are inadequately understood. We identify Fmn2, a non-diaphanous-related formin associated with cognitive disabilities, as a novel regulator of cooperative actin-microtubule remodelling in growth cones of both chick and zebrafish neurons. We show that Fmn2 stabilizes microtubules in the growth cones of cultured spinal neurons and in vivo. Super-resolution imaging revealed that Fmn2 facilitates guidance of exploratory microtubules along actin bundles into the chemosensory filopodia. Using live imaging, biochemistry and single-molecule assays, we show that a C-terminal domain in Fmn2 is necessary for the dynamic association between microtubules and actin filaments. In the absence of the cross-bridging function of Fmn2, filopodial capture of microtubules is compromised, resulting in destabilized filopodial protrusions and deficits in growth cone chemotaxis. Our results uncover a critical function for Fmn2 in actin-microtubule crosstalk in neurons and demonstrate that the modulation of microtubule dynamics via associations with F-actin is central to directional motility.
Collapse
Affiliation(s)
- Tanushree Kundu
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pune 411008, India
| | - Priyanka Dutta
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pune 411008, India
| | - Dhriti Nagar
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pune 411008, India
| | - Sankar Maiti
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, Nadia, West Bengal, India
| | - Aurnab Ghose
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pune 411008, India
| |
Collapse
|
32
|
Hou X, Nozumi M, Nakamura H, Igarashi M, Sugiyama S. Coactosin Promotes F-Actin Protrusion in Growth Cones Under Cofilin-Related Signaling Pathway. Front Cell Dev Biol 2021; 9:660349. [PMID: 34235144 PMCID: PMC8256272 DOI: 10.3389/fcell.2021.660349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/31/2021] [Indexed: 12/03/2022] Open
Abstract
During brain development, axon outgrowth and its subsequent pathfinding are reliant on a highly motile growth cone located at the tip of the axon. Actin polymerization that is regulated by actin-depolymerizing factors homology (ADF-H) domain-containing family drives the formation of lamellipodia and filopodia at the leading edge of growth cones for axon guidance. However, the precise localization and function of ADF-H domain-containing proteins involved in axon extension and retraction remain unclear. We have previously shown that transcripts and proteins of coactosin-like protein 1 (COTL1), an ADF-H domain-containing protein, are observed in neurites and axons in chick embryos. Coactosin overexpression analysis revealed that this protein was localized to axonal growth cones and involved in axon extension in the midbrain. We further examined the specific distribution of coactosin and cofilin within the growth cone using superresolution microscopy, structured illumination microscopy, which overcomes the optical diffraction limitation and is suitable to the analysis of cellular dynamic movements. We found that coactosin was tightly associated with F-actin bundles at the growth cones and that coactosin overexpression promoted the expansion of lamellipodia and extension of growth cones. Coactosin knockdown in oculomotor neurons resulted in an increase in the levels of the inactive, phosphorylated form of cofilin and dysregulation of actin polymerization and axonal elongation, which suggests that coactosin promoted axonal growth in a cofilin-dependent manner. Indeed, the application of a dominant-negative form of LIMK1, a downstream effector of GTPases, reversed the effect of coactosin knockdown on axonal growth by enhancing cofilin activity. Combined, our results indicate that coactosin functions promote the assembly of protrusive actin filament arrays at the leading edge for growth cone motility.
Collapse
Affiliation(s)
- Xubin Hou
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Department of Molecular Neurobiology, Graduate School of Life Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Harukazu Nakamura
- Department of Molecular Neurobiology, Graduate School of Life Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Sayaka Sugiyama
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
33
|
Crawley O, Grill B. Autophagy in axonal and presynaptic development. Curr Opin Neurobiol 2021; 69:139-148. [PMID: 33940492 DOI: 10.1016/j.conb.2021.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 10/21/2022]
Abstract
The study of autophagy in the nervous system has predominantly centered on degeneration. Evidence is now cementing crucial roles for autophagy in neuronal development and growth, especially in axonal and presynaptic compartments. A picture is emerging that autophagy typically promotes the growth of axons and reduces presynaptic stability. Nonetheless, these are not rigid principles, and it remains unclear why autophagy does not always display these relationships during axonal and presynaptic development. Recent progress has identified mechanisms underlying spatiotemporal control of autophagy in neurons and begun to unravel how autophagy is integrated with other cellular processes, such as proteasomal degradation and axon guidance. Ultimately, understanding how autophagy is regulated and its role in the developing nervous system is key to comprehending how the nervous system assembles its stereotyped yet plastic configuration. It is also likely to inform how we think about neurodevelopmental disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Oliver Crawley
- Unidad de Neurobiología Celular y de Sistemas, Instituto de Neurociencias (CSIC-UMH), San Juan de Alicante, 03550, Spain.
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98199, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
34
|
Camblor-Perujo S, Kononenko NL. Brain-specific functions of the endocytic machinery. FEBS J 2021; 289:2219-2246. [PMID: 33896112 DOI: 10.1111/febs.15897] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis is an essential cellular process required for multiple physiological functions, including communication with the extracellular environment, nutrient uptake, and signaling by the cell surface receptors. In a broad sense, endocytosis is accomplished through either constitutive or ligand-induced invagination of the plasma membrane, which results in the formation of the plasma membrane-retrieved endocytic vesicles, which can either be sent for degradation to the lysosomes or recycled back to the PM. This additional function of endocytosis in membrane retrieval has been adopted by excitable cells, such as neurons, for membrane equilibrium maintenance at synapses. The last two decades were especially productive with respect to the identification of brain-specific functions of the endocytic machinery, which additionally include but not limited to regulation of neuronal differentiation and migration, maintenance of neuron morphology and synaptic plasticity, and prevention of neurotoxic aggregates spreading. In this review, we highlight the current knowledge of brain-specific functions of endocytic machinery with a specific focus on three brain cell types, neuronal progenitor cells, neurons, and glial cells.
Collapse
Affiliation(s)
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, University of Cologne, Germany.,Center for Physiology & Pathophysiology, Medical Faculty, University of Cologne, Germany
| |
Collapse
|
35
|
Schneider F, Duong TA, Metz I, Winkelmeier J, Hübner CA, Endesfelder U, Rust MB. Mutual functional dependence of cyclase-associated protein 1 (CAP1) and cofilin1 in neuronal actin dynamics and growth cone function. Prog Neurobiol 2021; 202:102050. [PMID: 33845164 DOI: 10.1016/j.pneurobio.2021.102050] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/14/2021] [Accepted: 04/07/2021] [Indexed: 01/20/2023]
Abstract
Neuron connectivity depends on growth cones that navigate axons through the developing brain. Growth cones protrude and retract actin-rich structures to sense guidance cues. These cues control local actin dynamics and steer growth cones towards attractants and away from repellents, thereby directing axon outgrowth. Hence, actin binding proteins (ABPs) moved into the focus as critical regulators of neuron connectivity. We found cyclase-associated protein 1 (CAP1), an ABP with unknown brain function, abundant in growth cones. Super-resolution microscopy and live cell imaging combined with pharmacological approaches on hippocampal neurons from gene-targeted mice revealed a crucial role for CAP1 in actin dynamics that is critical for growth cone morphology and function. Growth cone defects in CAP1 knockout (KO) neurons compromised neuron differentiation and was associated with impaired neuron connectivity in CAP1-KO brains. Mechanistically, by rescue experiments in double KO neurons lacking CAP1 and the key actin regulator cofilin1, we demonstrated that CAP1 was essential for cofilin1 function in growth cone actin dynamics and morphology and vice versa. Together, we identified CAP1 as a novel actin regulator in growth cones that was relevant for neuron connectivity, and we demonstrated functional interdependence of CAP1 and cofilin1 in neuronal actin dynamics and growth cone function.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, 35032, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany; DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-University of Marburg, 35032, Marburg, Germany
| | - Thuy-An Duong
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, 35032, Marburg, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, 35032, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Jannik Winkelmeier
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology and LOEWE Center for Synthetic Microbiology (SYNMIKRO), 35043, Marburg, Germany; Department of Physics, Mellon College of Science, Carnegie-Mellon University, Pittsburgh, PA, USA
| | - Christian A Hübner
- Institute of Human Genetics, University Hospital Jena, 07743, Jena, Germany
| | - Ulrike Endesfelder
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology and LOEWE Center for Synthetic Microbiology (SYNMIKRO), 35043, Marburg, Germany; Department of Physics, Mellon College of Science, Carnegie-Mellon University, Pittsburgh, PA, USA
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, 35032, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany; DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-University of Marburg, 35032, Marburg, Germany.
| |
Collapse
|
36
|
Petrović A, Ban J, Tomljanović I, Pongrac M, Ivaničić M, Mikašinović S, Mladinic M. Establishment of Long-Term Primary Cortical Neuronal Cultures From Neonatal Opossum Monodelphis domestica. Front Cell Neurosci 2021; 15:661492. [PMID: 33815068 PMCID: PMC8012671 DOI: 10.3389/fncel.2021.661492] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/25/2021] [Indexed: 11/13/2022] Open
Abstract
Primary dissociated neuronal cultures have become a standard model for studying central nervous system (CNS) development. Such cultures are predominantly prepared from the hippocampus or cortex of rodents (mice and rats), while other mammals are less used. Here, we describe the establishment and extensive characterization of the primary dissociated neuronal cultures derived from the cortex of the gray South American short-tailed opossums, Monodelphis domestica. Opossums are unique in their ability to fully regenerate their CNS after an injury during their early postnatal development. Thus, we used cortex of postnatal day (P) 3-5 opossum to establish long-surviving and nearly pure neuronal cultures, as well as mixed cultures composed of radial glia cells (RGCs) in which their neurogenic and gliogenic potential was confirmed. Both types of cultures can survive for more than 1 month in vitro. We also prepared neuronal cultures from the P16-18 opossum cortex, which were composed of astrocytes and microglia, in addition to neurons. The long-surviving opossum primary dissociated neuronal cultures represent a novel mammalian in vitro platform particularly useful to study CNS development and regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Miranda Mladinic
- Laboratory for Molecular Neurobiology, Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
37
|
Meza U, Romero-Méndez C, Sánchez-Armáss S, Rodríguez-Menchaca AA. Role of rafts in neurological disorders. Neurologia 2021; 38:S0213-4853(21)00024-4. [PMID: 33726969 DOI: 10.1016/j.nrl.2021.01.008] [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: 09/03/2020] [Revised: 11/12/2020] [Accepted: 01/01/2021] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Rafts are function-structural cell membrane nano-domains. They contribute to explain the efficiency of signal transduction at the low physiological membrane concentrations of the signaling partners by their clustering inside specialized signaling domains. DEVELOPMENT In this article, we review the current model of the membrane rafts and their physio-pathological relevance in the nervous system, including their role in Parkinson, Alzheimer, and Huntington diseases. CONCLUSIONS Rafts disruption/dysfunction has been shown to relate diverse neurological diseases. Therefore, it has been suggested that preservation of membrane rafts may represent a strategy to prevent or delay neuronal dysfunctions in several diseases.
Collapse
Affiliation(s)
- U Meza
- Departamento de Fisiología y Biofísica. Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - C Romero-Méndez
- Departamento de Fisiología y Biofísica. Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - S Sánchez-Armáss
- Departamento de Fisiología y Biofísica. Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - A A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica. Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| |
Collapse
|
38
|
Pinto-Costa R, Sousa MM. Microtubules, actin and cytolinkers: how to connect cytoskeletons in the neuronal growth cone. Neurosci Lett 2021; 747:135693. [PMID: 33529653 DOI: 10.1016/j.neulet.2021.135693] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 01/20/2023]
Abstract
Cytolinkers ensure the integration of the different cytoskeleton components in the neuronal growth cone during development and in the course of axon regeneration. In neurons, an integrated skeleton guarantees appropriate function, and connectivity of high order circuits. Over the past years, several cytoskeleton regulatory proteins with actin-microtubule crosslinking activity have been identified. In neurons, the importance of spectrins, formins and other cytolinkers capable of coupling actin and microtubules has been extensively highlighted during axon outgrowth and guidance. In this Review, we discuss the current knowledge on cytolinkers specifically expressed in the neuronal growth cone of developing and regenerating axons.
Collapse
Affiliation(s)
- Rita Pinto-Costa
- Nerve Regeneration Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
| | - Monica Mendes Sousa
- Nerve Regeneration Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
39
|
Pollitt SL, Myers KR, Yoo J, Zheng JQ. LIM and SH3 protein 1 localizes to the leading edge of protruding lamellipodia and regulates axon development. Mol Biol Cell 2020; 31:2718-2732. [PMID: 32997597 PMCID: PMC7927181 DOI: 10.1091/mbc.e20-06-0366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The actin cytoskeleton drives cell motility and is essential for neuronal development and function. LIM and SH3 protein 1 (LASP1) is a unique actin-binding protein that is expressed in a wide range of cells including neurons, but its roles in cellular motility and neuronal development are not well understood. We report that LASP1 is expressed in rat hippocampus early in development, and this expression is maintained through adulthood. High-resolution imaging reveals that LASP1 is selectively concentrated at the leading edge of lamellipodia in migrating cells and axonal growth cones. This local enrichment of LASP1 is dynamically associated with the protrusive activity of lamellipodia, depends on the barbed ends of actin filaments, and requires both the LIM domain and the nebulin repeats of LASP1. Knockdown of LASP1 in cultured rat hippocampal neurons results in a substantial reduction in axonal outgrowth and arborization. Finally, loss of the Drosophila homologue Lasp from a subset of commissural neurons in the developing ventral nerve cord produces defasciculated axon bundles that do not reach their targets. Together, our data support a novel role for LASP1 in actin-based lamellipodial protrusion and establish LASP1 as a positive regulator of both in vitro and in vivo axon development.
Collapse
Affiliation(s)
| | | | - Jin Yoo
- Emory College, Emory University, Atlanta, GA 30322
| | - James Q Zheng
- Department of Cell Biology and.,Department of Neurology and Center for Neurodegenerative Diseases, Emory University School of Medicine, and
| |
Collapse
|
40
|
Kim MH, Park SR, Choi BH. Comparative Analysis of the Expression of Chondroitin Sulfate Subtypes and Their Inhibitory Effect on Axonal Growth in the Embryonic, Adult, and Injured Rat Brains. Tissue Eng Regen Med 2020; 18:165-178. [PMID: 32939673 DOI: 10.1007/s13770-020-00295-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/01/2020] [Accepted: 08/16/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Chondroitin sulfate glycosaminoglycans (CS-GAGs) are the primary inhibitory GAGs for neuronal growth after central nervous system (CNS) injury. However, the inhibitory or permissive activity of CS-GAG subtypes is controversial and depends on the physiological needs of CNS tissues. In this study, we investigated the characteristics and effects of CS-GAGs on axonal growth, which was isolated from the brain cortices of normal rat embryo at E18, normal adult rat brain and injured adult rat brain. METHODS Isolated CS-GAGs from embryo, normal adult, and injured adult rat brains were used for analyzing their effect on attachment and axonal growth using modified spot assay with dorsal root ganglion (DRG) explants and cerebellar granule neurons (CGNs). CS-GAGs were separated using high performance liquid chromatography (HPLC), and the subtypes of CS-GAGs were analyzed. RESULTS CS-GAGs of all three groups inhibited CGN attachment and axonal growth of DRGs. However, CS-GAGs of normal adult rat brain exhibited higher inhibitory activity than those of the other groups in both assays. When subtypes of CS-GAGs were analyzed using HPLC, CS-A (4S) was the most abundant in all three groups and found in largest amount in normal adult rat brain. In contrast, unsulfated CS (CS0) and CS-C (6S) were more abundant by 3-4-folds in E18 group than in the two adult groups. CONCLUSION When compared with the normal adult rat brain, injured rat brain showed relatively similar patterns to that of embryonic rat brain at E18 in the expression of CS subtypes and their inhibitory effect on axonal growth. This phenomenon could be due to differential expression of CS-GAGs subtypes causing decrease in the amount of CS-A and mature-type CS proteoglycan core proteins.
Collapse
Affiliation(s)
- Moon Hang Kim
- Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - So Ra Park
- Department of Physiology, Inha University College of Medicine, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Byung Hyune Choi
- Department of Biomedical Sciences, Inha University College of Medicine, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea.
| |
Collapse
|
41
|
Guo C, Webb SE, Chan CM, Miller AL. TPC2-mediated Ca 2+ signaling is required for axon extension in caudal primary motor neurons in zebrafish embryos. J Cell Sci 2020; 133:jcs244780. [PMID: 32546534 DOI: 10.1242/jcs.244780] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
The role of two-pore channel type 2 (TPC2, encoded by tcpn2)-mediated Ca2+ release was recently characterized in zebrafish during establishment of the early spinal circuitry, one of the key events in the coordination of neuromuscular activity. Here, we extend our study to investigate the in vivo role of TPC2 in the regulation of caudal primary motor neuron (CaP) axon extension. We used a combination of TPC2 knockdown with a translation-blocking morpholino antisense oligonucleotide (MO), TPC2 knockout via the generation of a tpcn2dhkz1a mutant line of zebrafish using CRISPR/Cas9 gene-editing and pharmacological inhibition of TPC2 via incubation with bafilomycin A1 (an H+-ATPase inhibitor) or trans-ned-19 (an NAADP receptor antagonist), and showed that these treatments attenuated CaP Ca2+ signaling and inhibited axon extension. We also characterized the expression of an arc1-like transcript in CaPs grown in primary culture. MO-mediated knockdown of ARC1-like in vivo led to attenuation of the Ca2+ transients in the CaP growth cones and an inhibition of axon extension. Together, our new data suggest a link between ARC1-like, TPC2 and Ca2+ signaling during axon extension in zebrafish.
Collapse
Affiliation(s)
- Chenxi Guo
- Division of Life Science and State Key Laboratory for Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Sarah E Webb
- Division of Life Science and State Key Laboratory for Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ching Man Chan
- Division of Life Science and State Key Laboratory for Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Andrew L Miller
- Division of Life Science and State Key Laboratory for Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
42
|
Nestin Selectively Facilitates the Phosphorylation of the Lissencephaly-Linked Protein Doublecortin (DCX) by cdk5/p35 to Regulate Growth Cone Morphology and Sema3a Sensitivity in Developing Neurons. J Neurosci 2020; 40:3720-3740. [PMID: 32273484 DOI: 10.1523/jneurosci.2471-19.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/24/2020] [Accepted: 03/24/2020] [Indexed: 11/21/2022] Open
Abstract
Nestin, an intermediate filament protein widely used as a marker of neural progenitors, was recently found to be expressed transiently in developing cortical neurons in culture and in developing mouse cortex. In young cortical cultures, nestin regulates axonal growth cone morphology. In addition, nestin, which is known to bind the neuronal cdk5/p35 kinase, affects responses to axon guidance cues upstream of cdk5, specifically, to Sema3a. Changes in growth cone morphology require rearrangements of cytoskeletal networks, and changes in microtubules and actin filaments are well studied. In contrast, the roles of intermediate filament proteins in this process are poorly understood, even in cultured neurons. Here, we investigate the molecular mechanism by which nestin affects growth cone morphology and Sema3a sensitivity. We find that nestin selectively facilitates the phosphorylation of the lissencephaly-linked protein doublecortin (DCX) by cdk5/p35, but the phosphorylation of other cdk5 substrates is not affected by nestin. We uncover that this substrate selectivity is based on the ability of nestin to interact with DCX, but not with other cdk5 substrates. Nestin thus creates a selective scaffold for DCX with activated cdk5/p35. Last, we use cortical cultures derived from Dcx KO mice to show that the effects of nestin on growth cone morphology and on Sema3a sensitivity are DCX-dependent, thus suggesting a functional role for the DCX-nestin complex in neurons. We propose that nestin changes growth cone behavior by regulating the intracellular kinase signaling environment in developing neurons. The sex of animal subjects is unknown.SIGNIFICANCE STATEMENT Nestin, an intermediate filament protein highly expressed in neural progenitors, was recently identified in developing neurons where it regulates growth cone morphology and responsiveness to the guidance cue Sema3a. Changes in growth cone morphology require rearrangements of cytoskeletal networks, but the roles of intermediate filaments in this process are poorly understood. We now report that nestin selectively facilitates phosphorylation of the lissencephaly-linked doublecortin (DCX) by cdk5/p35, but the phosphorylation of other cdk5 substrates is not affected. This substrate selectivity is based on preferential scaffolding of DCX, cdk5, and p35 by nestin. Additionally, we demonstrate a functional role for the DCX-nestin complex in neurons. We propose that nestin changes growth cone behavior by regulating intracellular kinase signaling in developing neurons.
Collapse
|
43
|
Dos-Santos Carvalho S, Moreau MM, Hien YE, Garcia M, Aubailly N, Henderson DJ, Studer V, Sans N, Thoumine O, Montcouquiol M. Vangl2 acts at the interface between actin and N-cadherin to modulate mammalian neuronal outgrowth. eLife 2020; 9:51822. [PMID: 31909712 PMCID: PMC6946565 DOI: 10.7554/elife.51822] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Dynamic mechanical interactions between adhesion complexes and the cytoskeleton are essential for axon outgrowth and guidance. Whether planar cell polarity (PCP) proteins, which regulate cytoskeleton dynamics and appear necessary for some axon guidance, also mediate interactions with membrane adhesion is still unclear. Here we show that Vangl2 controls growth cone velocity by regulating the internal retrograde actin flow in an N-cadherin-dependent fashion. Single molecule tracking experiments show that the loss of Vangl2 decreased fast-diffusing N-cadherin membrane molecules and increased confined N-cadherin trajectories. Using optically manipulated N-cadherin-coated microspheres, we correlated this behavior to a stronger mechanical coupling of N-cadherin with the actin cytoskeleton. Lastly, we show that the spatial distribution of Vangl2 within the growth cone is selectively affected by an N-cadherin-coated substrate. Altogether, our data show that Vangl2 acts as a negative regulator of axonal outgrowth by regulating the strength of the molecular clutch between N-cadherin and the actin cytoskeleton.
Collapse
Affiliation(s)
- Steve Dos-Santos Carvalho
- INSERM, Neurocentre Magendie, U1215, Bordeaux, France.,Univ. Bordeaux, Neurocentre Magendie, U1215, Bordeaux, France
| | - Maite M Moreau
- INSERM, Neurocentre Magendie, U1215, Bordeaux, France.,Univ. Bordeaux, Neurocentre Magendie, U1215, Bordeaux, France
| | - Yeri Esther Hien
- INSERM, Neurocentre Magendie, U1215, Bordeaux, France.,Univ. Bordeaux, Neurocentre Magendie, U1215, Bordeaux, France
| | - Mikael Garcia
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France.,Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Nathalie Aubailly
- INSERM, Neurocentre Magendie, U1215, Bordeaux, France.,Univ. Bordeaux, Neurocentre Magendie, U1215, Bordeaux, France
| | - Deborah J Henderson
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne, United Kingdom
| | - Vincent Studer
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France.,Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Nathalie Sans
- INSERM, Neurocentre Magendie, U1215, Bordeaux, France.,Univ. Bordeaux, Neurocentre Magendie, U1215, Bordeaux, France
| | - Olivier Thoumine
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France.,Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Mireille Montcouquiol
- INSERM, Neurocentre Magendie, U1215, Bordeaux, France.,Univ. Bordeaux, Neurocentre Magendie, U1215, Bordeaux, France
| |
Collapse
|
44
|
Guo J, Otis JM, Suciu SK, Catalano C, Xing L, Constable S, Wachten D, Gupton S, Lee J, Lee A, Blackley KH, Ptacek T, Simon JM, Schurmans S, Stuber GD, Caspary T, Anton ES. Primary Cilia Signaling Promotes Axonal Tract Development and Is Disrupted in Joubert Syndrome-Related Disorders Models. Dev Cell 2019; 51:759-774.e5. [PMID: 31846650 PMCID: PMC6953258 DOI: 10.1016/j.devcel.2019.11.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/08/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022]
Abstract
Appropriate axonal growth and connectivity are essential for functional wiring of the brain. Joubert syndrome-related disorders (JSRD), a group of ciliopathies in which mutations disrupt primary cilia function, are characterized by axonal tract malformations. However, little is known about how cilia-driven signaling regulates axonal growth and connectivity. We demonstrate that the deletion of related JSRD genes, Arl13b and Inpp5e, in projection neurons leads to de-fasciculated and misoriented axonal tracts. Arl13b deletion disrupts the function of its downstream effector, Inpp5e, and deregulates ciliary-PI3K/AKT signaling. Chemogenetic activation of ciliary GPCR signaling and cilia-specific optogenetic modulation of downstream second messenger cascades (PI3K, AKT, and AC3) commonly regulated by ciliary signaling receptors induce rapid changes in axonal dynamics. Further, Arl13b deletion leads to changes in transcriptional landscape associated with dysregulated PI3K/AKT signaling. These data suggest that ciliary signaling acts to modulate axonal connectivity and that impaired primary cilia signaling underlies axonal tract defects in JSRD.
Collapse
Affiliation(s)
- Jiami Guo
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA.
| | - James M Otis
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sarah K Suciu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christy Catalano
- Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA
| | - Lei Xing
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sandii Constable
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dagmar Wachten
- Biophysical Imaging, Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Stephanie Gupton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Janice Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Amelia Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Katherine H Blackley
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Travis Ptacek
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Stephane Schurmans
- Laboratory of Functional Genetics, GIGA Research Center, University of Liège, Liège, Belgium
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain and Emotion, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
45
|
Thomason EJ, Escalante M, Osterhout DJ, Fuss B. The oligodendrocyte growth cone and its actin cytoskeleton: A fundamental element for progenitor cell migration and CNS myelination. Glia 2019; 68:1329-1346. [PMID: 31696982 DOI: 10.1002/glia.23735] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/26/2019] [Accepted: 10/01/2019] [Indexed: 01/06/2023]
Abstract
Cells of the oligodendrocyte (OLG) lineage engage in highly motile behaviors that are crucial for effective central nervous system (CNS) myelination. These behaviors include the guided migration of OLG progenitor cells (OPCs), the surveying of local environments by cellular processes extending from differentiating and pre-myelinating OLGs, and during the process of active myelin wrapping, the forward movement of the leading edge of the myelin sheath's inner tongue along the axon. Almost all of these motile behaviors are driven by actin cytoskeletal dynamics initiated within a lamellipodial structure that is located at the tip of cellular OLG/OPC processes and is structurally as well as functionally similar to the neuronal growth cone. Accordingly, coordinated stoichiometries of actin filament (F-actin) assembly and disassembly at these OLG/OPC growth cones have been implicated in directing process outgrowth and guidance, and the initiation of myelination. Nonetheless, the functional importance of the OLG/OPC growth cone still remains to be fully understood, and, as a unique aspect of actin cytoskeletal dynamics, F-actin depolymerization and disassembly start to predominate at the transition from myelination initiation to myelin wrapping. This review provides an overview of the current knowledge about OLG/OPC growth cones, and it proposes a model in which actin cytoskeletal dynamics in OLG/OPC growth cones are a main driver for morphological transformations and motile behaviors. Remarkably, these activities, at least at the later stages of OLG maturation, may be regulated independently from the transcriptional gene expression changes typically associated with CNS myelination.
Collapse
Affiliation(s)
- Elizabeth J Thomason
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Miguel Escalante
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia.,Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Donna J Osterhout
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
46
|
Chighizola M, Dini T, Lenardi C, Milani P, Podestà A, Schulte C. Mechanotransduction in neuronal cell development and functioning. Biophys Rev 2019; 11:701-720. [PMID: 31617079 PMCID: PMC6815321 DOI: 10.1007/s12551-019-00587-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/29/2019] [Indexed: 12/21/2022] Open
Abstract
Although many details remain still elusive, it became increasingly evident in recent years that mechanosensing of microenvironmental biophysical cues and subsequent mechanotransduction are strongly involved in the regulation of neuronal cell development and functioning. This review gives an overview about the current understanding of brain and neuronal cell mechanobiology and how it impacts on neurogenesis, neuronal migration, differentiation, and maturation. We will focus particularly on the events in the cell/microenvironment interface and the decisive extracellular matrix (ECM) parameters (i.e. rigidity and nanometric spatial organisation of adhesion sites) that modulate integrin adhesion complex-based mechanosensing and mechanotransductive signalling. It will also be outlined how biomaterial approaches mimicking essential ECM features help to understand these processes and how they can be used to control and guide neuronal cell behaviour by providing appropriate biophysical cues. In addition, principal biophysical methods will be highlighted that have been crucial for the study of neuronal mechanobiology.
Collapse
Affiliation(s)
- Matteo Chighizola
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Tania Dini
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Cristina Lenardi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Paolo Milani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Alessandro Podestà
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Carsten Schulte
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy.
| |
Collapse
|
47
|
Brettle M, Stefen H, Djordjevic A, Fok SYY, Chan JW, van Hummel A, van der Hoven J, Przybyla M, Volkerling A, Ke YD, Delerue F, Ittner LM, Fath T. Developmental Expression of Mutant PFN1 in Motor Neurons Impacts Neuronal Growth and Motor Performance of Young and Adult Mice. Front Mol Neurosci 2019; 12:231. [PMID: 31611772 PMCID: PMC6776973 DOI: 10.3389/fnmol.2019.00231] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease with limited treatment and no cure. Mutations in profilin 1 were identified as a cause of familial ALS (fALS) in 2012. We investigated the functional impact of mutant profilin 1 expression in spinal cords during mouse development. We developed a novel mouse model with the expression of profilin 1 C71G under the control of the Hb9 promoter, targeting expression to α-motor neurons in the spinal cord during development. Embryos of transgenic mice showed evidence of a significant reduction of brachial nerve diameter and a loss of Mendelian inheritance. Despite the lack of transgene expression, adult mice presented with significant motor deficits. Transgenic mice had a significant reduction in the number of motor neurons in the spinal cord. Further analysis of these motor neurons in aged transgenic mice revealed reduced levels of TDP-43 and ChAT expression. Although profilin 1 C71G was only expressed during development, adult mice presented with some ALS-associated pathology and motor symptoms. This study highlights the effect of profilin 1 during neurodevelopment and the impact that this may have in later ALS.
Collapse
Affiliation(s)
- Merryn Brettle
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Randwick, NSW, Australia.,Biomedical Imaging Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Randwick, NSW, Australia
| | - Holly Stefen
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Aleksandra Djordjevic
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Randwick, NSW, Australia
| | - Sandra Y Y Fok
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Randwick, NSW, Australia.,Biomedical Imaging Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Randwick, NSW, Australia
| | - Josephine W Chan
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Randwick, NSW, Australia
| | - Annika van Hummel
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Julia van der Hoven
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Magdalena Przybyla
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Alexander Volkerling
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Randwick, NSW, Australia
| | - Yazi D Ke
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Fabien Delerue
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Lars M Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Thomas Fath
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Randwick, NSW, Australia.,Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
48
|
Bodakuntla S, Jijumon AS, Villablanca C, Gonzalez-Billault C, Janke C. Microtubule-Associated Proteins: Structuring the Cytoskeleton. Trends Cell Biol 2019; 29:804-819. [PMID: 31416684 DOI: 10.1016/j.tcb.2019.07.004] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 11/27/2022]
Abstract
Microtubule-associated proteins (MAPs) were initially discovered as proteins that bind to and stabilize microtubules. Today, an ever-growing number of MAPs reveals a more complex picture of these proteins as organizers of the microtubule cytoskeleton that have a large variety of functions. MAPs enable microtubules to participate in a plethora of cellular processes such as the assembly of mitotic and meiotic spindles, neuronal development, and the formation of the ciliary axoneme. Although some subgroups of MAPs have been exhaustively characterized, a strikingly large number of MAPs remain barely characterized other than their interactions with microtubules. We provide a comprehensive view on the currently known MAPs in mammals. We discuss their molecular mechanisms and functions, as well as their physiological role and links to pathologies.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 3348, F-91405 Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, F-91405 Orsay, France
| | - A S Jijumon
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 3348, F-91405 Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, F-91405 Orsay, France
| | - Cristopher Villablanca
- Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Christian Gonzalez-Billault
- Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile.
| | - Carsten Janke
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 3348, F-91405 Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, F-91405 Orsay, France.
| |
Collapse
|
49
|
Tsujioka H, Yamashita T. Comparison of gene expression profile of the spinal cord of sprouting-capable neonatal and sprouting-incapable adult mice. BMC Genomics 2019; 20:619. [PMID: 31362699 PMCID: PMC6668129 DOI: 10.1186/s12864-019-5974-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
Background The regenerative ability of severed axons in the central nervous system is limited in mammals. However, after central nervous system injury, neural function is partially recovered by the formation of a compensatory neural circuit. In a mouse pyramidotomy model, axonal sprouting of the intact side of the corticospinal tract is observed in the spinal cord, and the axons make new synapses with the denervated side of propriospinal neurons. Moreover, this sprouting ability is enhanced in neonatal mice compared to that in adult mice. Myelin-associated molecules in the spinal cord or intrinsic factors in corticospinal neurons have been investigated in previous studies, but the factors that determine elevated sprouting ability in neonatal mice are not fully understood. Further, in the early phase after pyramidotomy, glial responses are observed in the spinal cord. To elucidate the basal difference in the spinal cord, we compared gene expression profiles of entire C4–7 cervical cord tissues between neonatal (injured at postnatal day 7) and adult (injured at 8 weeks of age) mice by RNA-sequencing. We also tried to identify discordant gene expression changes that might inhibit axonal sprouting in adult mice at the early phase (3 days) after pyramidotomy. Results A comparison of neonatal and adult sham groups revealed remarkable basal differences in the spinal cord, such as active neural circuit formation, cell proliferation, the development of myelination, and an immature immune system in neonatal mice compared to that observed in adult mice. Some inflammation-related genes were selectively expressed in adult mice after pyramidotomy, implying the possibility that these genes might be related to the low sprouting ability in adult mice. Conclusions This study provides useful information regarding the basal difference between neonatal and adult spinal cords and the possible differential response after pyramidotomy, both of which are necessary to understand why sprouting ability is increased in neonatal mice compared to that in adult mice. Electronic supplementary material The online version of this article (10.1186/s12864-019-5974-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hiroshi Tsujioka
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan. .,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan. .,Graduate School of Frontier Bioscience, Osaka University, Osaka, Japan. .,Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
50
|
Shao Q, Yang T, Huang H, Majumder T, Khot BA, Khouzani MM, Alarmanazi F, Gore YK, Liu G. Disease-associated mutations in human TUBB3 disturb netrin repulsive signaling. PLoS One 2019; 14:e0218811. [PMID: 31226147 PMCID: PMC6588280 DOI: 10.1371/journal.pone.0218811] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/10/2019] [Indexed: 11/28/2022] Open
Abstract
Missense mutations in the human TUBB3 gene cause a variety of neurological disorders associated with defects in axon guidance and neuronal migration, but the underlying molecular mechanisms are not well understood. Recent studies have shown that direct coupling of dynamic TUBB3 in microtubules with netrin receptors is required for netrin-1-mediated axon guidance, and the interaction of netrin-1 repulsive receptor UNC5C with TUBB3 is involved in netrin-1 mediated axonal repulsion. Here, we report that TUBB3 mutations perturb netrin-1/UNC5C repulsive signaling in the developing nervous system. Among twelve mutants reported in previous studies, five of them show significantly reduced interaction with UNC5C in comparison to the wild-type TUBB3. TUBB3 mutants R262C and R62Q exhibit decreased subcellular colocalization with UNC5C in the peripheral area of the growth cone of primary mouse neurons. Netrin-1 reduces the colocalization of UNC5C with wild-type TUBB3, but not TUBB3 mutants R262C or R62Q, in the growth cone. Results from the in vitro cosedimentation assay indicate that netrin-1 inhibits cosedimentation of UNC5C with polymerized microtubules in primary mouse neurons expressing the wild-type TUBB3, but not R262C or R62Q. Expression of either R262C or R62Q not only blocks netrin-1-induced growth cone collapse and axonal repulsion of primary EGL cells in vitro, but also results in axon projections defects of chicken dorsal root ganglion neurons in ovo. Our study reveals that human TUBB3 mutations specifically perturb netrin-1/UNC5C-mediated repulsion.
Collapse
Affiliation(s)
- Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Huai Huang
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Bhakti Ajit Khot
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | | | - Farrah Alarmanazi
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Yasmin K. Gore
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
- * E-mail:
| |
Collapse
|