1
|
Zupančič M, Keimpema E, Tretiakov EO, Eder SJ, Lev I, Englmaier L, Bhandari P, Fietz SA, Härtig W, Renaux E, Villunger A, Hökfelt T, Zimmer M, Clotman F, Harkany T. Concerted transcriptional regulation of the morphogenesis of hypothalamic neurons by ONECUT3. Nat Commun 2024; 15:8631. [PMID: 39366958 PMCID: PMC11452682 DOI: 10.1038/s41467-024-52762-z] [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: 12/02/2022] [Accepted: 09/19/2024] [Indexed: 10/06/2024] Open
Abstract
Acquisition of specialized cellular features is controlled by the ordered expression of transcription factors (TFs) along differentiation trajectories. Here, we find a member of the Onecut TF family, ONECUT3, expressed in postmitotic neurons that leave their Ascl1+/Onecut1/2+ proliferative domain in the vertebrate hypothalamus to instruct neuronal differentiation. We combined single-cell RNA-seq and gain-of-function experiments for gene network reconstruction to show that ONECUT3 affects the polarization and morphogenesis of both hypothalamic GABA-derived dopamine and thyrotropin-releasing hormone (TRH)+ glutamate neurons through neuron navigator-2 (NAV2). In vivo, siRNA-mediated knockdown of ONECUT3 in neonatal mice reduced NAV2 mRNA, as well as neurite complexity in Onecut3-containing neurons, while genetic deletion of Onecut3/ceh-48 in C. elegans impaired neurocircuit wiring, and sensory discrimination-based behaviors. Thus, ONECUT3, conserved across neuronal subtypes and many species, underpins the polarization and morphological plasticity of phenotypically distinct neurons that descend from a common pool of Ascl1+ progenitors in the hypothalamus.
Collapse
Affiliation(s)
- Maja Zupančič
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | - Evgenii O Tretiakov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Stephanie J Eder
- Department of Neuroscience and Developmental Biology, Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
- Vienna Biocenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Itamar Lev
- Department of Neuroscience and Developmental Biology, Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Lukas Englmaier
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Pradeep Bhandari
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Simone A Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Estelle Renaux
- Animal Molecular and Cellular Biology, Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Andreas Villunger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden
| | - Manuel Zimmer
- Department of Neuroscience and Developmental Biology, Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Frédéric Clotman
- Animal Molecular and Cellular Biology, Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
2
|
Eid L, Lokmane L, Raju PK, Tene Tadoum SB, Jiang X, Toulouse K, Lupien-Meilleur A, Charron-Ligez F, Toumi A, Backer S, Lachance M, Lavertu-Jolin M, Montseny M, Lacaille JC, Bloch-Gallego E, Rossignol E. Both GEF domains of the autism and developmental epileptic encephalopathy-associated Trio protein are required for proper tangential migration of GABAergic interneurons. Mol Psychiatry 2024:10.1038/s41380-024-02742-y. [PMID: 39300136 DOI: 10.1038/s41380-024-02742-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
Recessive and de novo mutations in the TRIO gene are associated with intellectual deficiency (ID), autism spectrum disorder (ASD) and developmental epileptic encephalopathies (DEE). TRIO is a dual guanine nucleotide exchange factor (GEF) that activates Rac1, Cdc42 and RhoA. Trio has been extensively studied in excitatory neurons, and has recently been found to regulate the switch from tangential to radial migration in GABAergic interneurons (INs) through GEFD1-Rac1-dependent SDF1α/CXCR4 signaling. Given the central role of Rho-GTPases during neuronal migration and the implication of IN pathologies in ASD and DEE, we investigated the relative roles of both Trio's GEF domains in regulating the dynamics of INs tangential migration. In Trio-/- mice, we observed reduced numbers of tangentially migrating INs, with intact progenitor proliferation. Further, we noted increased growth cone collapse in developing INs, suggesting altered cytoskeleton dynamics. To bypass the embryonic mortality of Trio-/- mice, we generated Dlx5/6Cre;Trioc/c conditional mutant mice (TriocKO), which develop spontaneous seizures and behavioral deficits reminiscent of ASD and ID. These phenotypes are associated with reduced cortical IN density and functional cortical inhibition. Mechanistically, this reduction of cortical IN numbers reflects a premature switch to radial migration, with an aberrant early entry in the cortical plate, as well as major deficits in cytoskeletal dynamics, including enhanced leading neurite branching and slower nucleokinesis reflecting reduced actin filament condensation and turnover as well as a loss of response to the motogenic effect of EphA4/ephrin A2 reverse signaling. Further, we show that both Trio GEFD1 and GEFD2 domains are required for proper IN migration, with a dominant role of the RhoA-activating GEFD2 domain. Altogether, our data show a critical role of the DEE/ASD-associated Trio gene in the establishment of cortical inhibition and the requirement of both GEF domains in regulating IN migration dynamics.
Collapse
Affiliation(s)
- Lara Eid
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
- Département de neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Ludmilla Lokmane
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Praveen K Raju
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
- Département de neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Samuel Boris Tene Tadoum
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
- Département de neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Xiao Jiang
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Karolanne Toulouse
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
- Département de neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Alexis Lupien-Meilleur
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
- Département de neurosciences, Université de Montréal, Montréal, QC, Canada
| | - François Charron-Ligez
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Asmaa Toumi
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Stéphanie Backer
- Institut Cochin- INSERM, U1016-CNRS UMR 8104-Université Paris Cité -24, rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Mathieu Lachance
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Marisol Lavertu-Jolin
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Marie Montseny
- Institut Cochin- INSERM, U1016-CNRS UMR 8104-Université Paris Cité -24, rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Jean-Claude Lacaille
- Département de neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre interdisciplinaire de recherche sur le cerveau et l'apprentissage, Groupe de recherche sur la signalisation neurale et la circuiterie, Université de Montréal, Montréal, QC, Canada
| | - Evelyne Bloch-Gallego
- Institut Cochin- INSERM, U1016-CNRS UMR 8104-Université Paris Cité -24, rue du Faubourg Saint-Jacques, 75014, Paris, France.
| | - Elsa Rossignol
- Centre de recherche du CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, H3T 1C5, Canada.
- Département de neurosciences, Université de Montréal, Montréal, QC, Canada.
- Département de Pédiatrie, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
3
|
Yang GN, Sun YBY, Roberts PK, Moka H, Sung MK, Gardner-Russell J, El Wazan L, Toussaint B, Kumar S, Machin H, Dusting GJ, Parfitt GJ, Davidson K, Chong EW, Brown KD, Polo JM, Daniell M. Exploring single-cell RNA sequencing as a decision-making tool in the clinical management of Fuchs' endothelial corneal dystrophy. Prog Retin Eye Res 2024; 102:101286. [PMID: 38969166 DOI: 10.1016/j.preteyeres.2024.101286] [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: 01/17/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has enabled the identification of novel gene signatures and cell heterogeneity in numerous tissues and diseases. Here we review the use of this technology for Fuchs' Endothelial Corneal Dystrophy (FECD). FECD is the most common indication for corneal endothelial transplantation worldwide. FECD is challenging to manage because it is genetically heterogenous, can be autosomal dominant or sporadic, and progress at different rates. Single-cell RNA sequencing has enabled the discovery of several FECD subtypes, each with associated gene signatures, and cell heterogeneity. Current FECD treatments are mainly surgical, with various Rho kinase (ROCK) inhibitors used to promote endothelial cell metabolism and proliferation following surgery. A range of emerging therapies for FECD including cell therapies, gene therapies, tissue engineered scaffolds, and pharmaceuticals are in preclinical and clinical trials. Unlike conventional disease management methods based on clinical presentations and family history, targeting FECD using scRNA-seq based precision-medicine has the potential to pinpoint the disease subtypes, mechanisms, stages, severities, and help clinicians in making the best decision for surgeries and the applications of therapeutics. In this review, we first discuss the feasibility and potential of using scRNA-seq in clinical diagnostics for FECD, highlight advances from the latest clinical treatments and emerging therapies for FECD, integrate scRNA-seq results and clinical notes from our FECD patients and discuss the potential of applying alternative therapies to manage these cases clinically.
Collapse
Affiliation(s)
- Gink N Yang
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Yu B Y Sun
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Philip Ke Roberts
- Department of Ophthalmology, Medical University Vienna, 18-20 Währinger Gürtel, Vienna, Austria
| | - Hothri Moka
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Min K Sung
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Jesse Gardner-Russell
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Layal El Wazan
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Bridget Toussaint
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Satheesh Kumar
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Heather Machin
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Lions Eye Donation Service, Level 7, Smorgon Family Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia
| | - Gregory J Dusting
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Geraint J Parfitt
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Kathryn Davidson
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Elaine W Chong
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Department of Ophthalmology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Karl D Brown
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Jose M Polo
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Mark Daniell
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Lions Eye Donation Service, Level 7, Smorgon Family Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Škarková A, Pelantová M, Tolde O, Legátová A, Mateu R, Bušek P, Garcia-Borja E, Šedo A, Etienne-Manneville S, Rösel D, Brábek J. Microtubule-associated NAV3 regulates invasive phenotypes in glioblastoma cells. Brain Pathol 2024:e13294. [PMID: 39097525 DOI: 10.1111/bpa.13294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/17/2024] [Indexed: 08/05/2024] Open
Abstract
Glioblastomas are aggressive brain tumors for which effective therapy is still lacking, resulting in dismal survival rates. These tumors display significant phenotypic plasticity, harboring diverse cell populations ranging from tumor core cells to dispersed, highly invasive cells. Neuron navigator 3 (NAV3), a microtubule-associated protein affecting microtubule growth and dynamics, is downregulated in various cancers, including glioblastoma, and has thus been considered a tumor suppressor. In this study, we challenge this designation and unveil distinct expression patterns of NAV3 across different invasion phenotypes. Using glioblastoma cell lines and patient-derived glioma stem-like cell cultures, we disclose an upregulation of NAV3 in invading glioblastoma cells, contrasting with its lower expression in cells residing in tumor spheroid cores. Furthermore, we establish an association between low and high NAV3 expression and the amoeboid and mesenchymal invasive phenotype, respectively, and demonstrate that overexpression of NAV3 directly stimulates glioblastoma invasive behavior in both 2D and 3D environments. Consistently, we observed increased NAV3 expression in cells migrating along blood vessels in mouse xenografts. Overall, our results shed light on the role of NAV3 in glioblastoma invasion, providing insights into this lethal aspect of glioblastoma behavior.
Collapse
Affiliation(s)
- Aneta Škarková
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Markéta Pelantová
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Ondřej Tolde
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Anna Legátová
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Rosana Mateu
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Bušek
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Elena Garcia-Borja
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Aleksi Šedo
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Université Paris Cité, UMR3691 CNRS, Institut Pasteur, Paris, France
| | - Daniel Rösel
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Jan Brábek
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| |
Collapse
|
5
|
Gossen S, Gerstner S, Borchers A. The RhoGEF Trio is transported by microtubules and affects microtubule stability in migrating neural crest cells. Cells Dev 2024; 177:203899. [PMID: 38160720 DOI: 10.1016/j.cdev.2023.203899] [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: 11/01/2023] [Revised: 12/08/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Directed cell migration requires a local fine-tuning of Rho GTPase activity to control protrusion formation, cell-cell contraction, and turnover of cellular adhesions. The Rho guanine nucleotide exchange factor (GEF) TRIO is ideally suited to control RhoGTPase activity because it combines two distinct catalytic domains to control Rac1 and RhoA activity in one molecule. However, at the cellular level, this molecular feature also requires a tight spatiotemporal control of TRIO activity. Here, we analyze the dynamic localization of Trio in Xenopus cranial neural crest (NC) cells, where we have recently shown that Trio is required for protrusion formation and migration. Using live cell imaging, we find that the GEF2 domain, but not the GEF1 domain of Trio, dynamically colocalizes with EB3 at microtubule plus-ends. Microtubule-mediated transport of Trio appears to be relevant for its function in NC migration, as a mutant GEF2 construct lacking the SxIP motif responsible for microtubule plus-end localization was significantly impaired in its ability to rescue the Trio loss-of-function phenotype compared to wild-type GEF2. Furthermore, by analyzing microtubule dynamics in migrating NC cells, we observed that loss of Trio function stabilized microtubules at cell-cell contact sites compared to controls, whereas they were destabilized at the leading edge of NC cells. Our data suggest that Trio is transported by microtubules to distinct subcellular locations where it has different functions in controlling microtubule stability, cell morphology, and cell-cell interaction during directed NC migration.
Collapse
Affiliation(s)
- Stefanie Gossen
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Karl-von-Frisch-Str. 8, 35043 Marburg, Germany
| | - Sarah Gerstner
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Karl-von-Frisch-Str. 8, 35043 Marburg, Germany
| | - Annette Borchers
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Karl-von-Frisch-Str. 8, 35043 Marburg, Germany.
| |
Collapse
|
6
|
Sandeep P, Sharma P, Luhach K, Dhiman N, Kharkwal H, Sharma B. Neuron navigators: A novel frontier with physiological and pathological implications. Mol Cell Neurosci 2023; 127:103905. [PMID: 37972804 DOI: 10.1016/j.mcn.2023.103905] [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: 09/12/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
Neuron navigators are microtubule plus-end tracking proteins containing basic and serine rich regions which are encoded by neuron navigator genes (NAVs). Neuron navigator proteins are essential for neurite outgrowth, neuronal migration, and overall neurodevelopment along with some other functions as well. The navigator proteins are substantially expressed in the developing brain and have been reported to be differentially expressed in various tissues at different ages. Over the years, the research has found neuron navigators to be implicated in a spectrum of pathological conditions such as developmental anomalies, neurodegenerative disorders, neuropathic pain, anxiety, cancers, and certain inflammatory conditions. The existing knowledge about neuron navigators remains sparse owing to their differential functions, undiscovered modulators, and unknown molecular mechanisms. Investigating the possible role of neuron navigators in various physiological processes and pathological conditions pose as a novel field that requires extensive research and might provide novel mechanistic insights and understanding of these aspects.
Collapse
Affiliation(s)
- Parth Sandeep
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Noida, India
| | - Poonam Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Noida, India
| | - Kanishk Luhach
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Noida, India
| | - Neerupma Dhiman
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Noida, India
| | - Harsha Kharkwal
- Amity Natural and Herbal Product Research, Amity Institute of Phytochemistry and Phytomedicine, Amity University, Uttar Pradesh, India
| | - Bhupesh Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Noida, India.
| |
Collapse
|
7
|
Bagley JR, Tan Y, Zhu W, Cheng Z, Takeda S, Fang Z, Arslan A, Wang M, Guan Y, Jiang L, Jian R, Gu F, Parada I, Prince D, Jentsch JD, Peltz G. Neuron Navigator 1 (Nav1) regulates the response to cocaine in mice. Commun Biol 2023; 6:1053. [PMID: 37853211 PMCID: PMC10584906 DOI: 10.1038/s42003-023-05430-9] [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: 05/15/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023] Open
Abstract
Genetic variation accounts for much of the risk for developing a substance use disorder, but the underlying genetic factors and their genetic effector mechanisms are mostly unknown. Inbred mouse strains exhibit substantial and heritable differences in the extent of voluntary cocaine self-administration. Computational genetic analysis of cocaine self-administration data obtained from twenty-one inbred strains identified Nav1, a member of the neuron navigator family that regulates dendrite formation and axonal guidance, as a candidate gene. To test this genetic hypothesis, we generated and characterized Nav1 knockout mice. Consistent with the genetic prediction, Nav1 knockout mice exhibited increased voluntary cocaine intake and had increased motivation for cocaine consumption. Immunohistochemistry, electrophysiology, and transcriptomic studies were performed as a starting point for investigating the mechanism for the Nav1 knockout effect. Nav1 knockout mice had a reduced inhibitory synapse density in their cortex, increased excitatory synaptic transmission in their cortex and hippocampus, and increased excitatory neurons in a deep cortical layer. Collectively, our results indicate that Nav1 regulates the response to cocaine, and we identified Nav1 knockout induced changes in the excitatory and inhibitory synaptic balance in the cortex and hippocampus that could contribute to this effect.
Collapse
Affiliation(s)
- Jared R Bagley
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Yalun Tan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Wan Zhu
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Zhuanfen Cheng
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Saori Takeda
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Zhouqing Fang
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Ahmed Arslan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Meiyue Wang
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Yuan Guan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University Medical School, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University Medical School, Stanford, CA, USA
| | - Feng Gu
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
- Department of Biological Sciences, University of North Texas, Denton, USA
| | - Isabel Parada
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
| | - David Prince
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
| | - J David Jentsch
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Gary Peltz
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA.
| |
Collapse
|
8
|
Toudji I, Toumi A, Chamberland É, Rossignol E. Interneuron odyssey: molecular mechanisms of tangential migration. Front Neural Circuits 2023; 17:1256455. [PMID: 37779671 PMCID: PMC10538647 DOI: 10.3389/fncir.2023.1256455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Cortical GABAergic interneurons are critical components of neural networks. They provide local and long-range inhibition and help coordinate network activities involved in various brain functions, including signal processing, learning, memory and adaptative responses. Disruption of cortical GABAergic interneuron migration thus induces profound deficits in neural network organization and function, and results in a variety of neurodevelopmental and neuropsychiatric disorders including epilepsy, intellectual disability, autism spectrum disorders and schizophrenia. It is thus of paramount importance to elucidate the specific mechanisms that govern the migration of interneurons to clarify some of the underlying disease mechanisms. GABAergic interneurons destined to populate the cortex arise from multipotent ventral progenitor cells located in the ganglionic eminences and pre-optic area. Post-mitotic interneurons exit their place of origin in the ventral forebrain and migrate dorsally using defined migratory streams to reach the cortical plate, which they enter through radial migration before dispersing to settle in their final laminar allocation. While migrating, cortical interneurons constantly change their morphology through the dynamic remodeling of actomyosin and microtubule cytoskeleton as they detect and integrate extracellular guidance cues generated by neuronal and non-neuronal sources distributed along their migratory routes. These processes ensure proper distribution of GABAergic interneurons across cortical areas and lamina, supporting the development of adequate network connectivity and brain function. This short review summarizes current knowledge on the cellular and molecular mechanisms controlling cortical GABAergic interneuron migration, with a focus on tangential migration, and addresses potential avenues for cell-based interneuron progenitor transplants in the treatment of neurodevelopmental disorders and epilepsy.
Collapse
Affiliation(s)
- Ikram Toudji
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Asmaa Toumi
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Émile Chamberland
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Elsa Rossignol
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
9
|
Wang W, Li W, Pan L, Li L, Xu Y, Wang Y, Zhang X, Zhang S. Dynamic Regulation Genes at Microtubule Plus Ends: A Novel Class of Glioma Biomarkers. BIOLOGY 2023; 12:biology12030488. [PMID: 36979179 PMCID: PMC10045452 DOI: 10.3390/biology12030488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023]
Abstract
Simple Summary Microtubule plus-end-related genes (MPERGs) encode a group of proteins that specifically aggregate at the microtubule plus ends to play critical biological roles in the cell cycle, cell movement, ciliogenesis, and neuronal development by coordinating microtubule assembly and dynamics; however, the MPERG correlations and their clinical significance in glioma are not fully understood. This study is the first to systematically analyze and define a seven-gene signature (CTTNBP2, KIF18A, NAV1, SLAIN2, SRCIN1, TRIO, and TTBK2) and nomogram model closely associated with clinical factors and the tumor microenvironment as a reliable and independent prognostic biomarker to guide personalized choices of immunotherapy and chemotherapy for glioma patients. Abstract Glioma is the most prevalent and aggressive primary nervous system tumor with an unfavorable prognosis. Microtubule plus-end-related genes (MPERGs) play critical biological roles in the cell cycle, cell movement, ciliogenesis, and neuronal development by coordinating microtubule assembly and dynamics. This research seeks to systematically explore the oncological characteristics of these genes in microtubule-enriched glioma, focusing on developing a novel MPERG-based prognostic signature to improve the prognosis and provide more treatment options for glioma patients. First, we thoroughly analyzed and identified 45 differentially expressed MPERGs in glioma. Based on these genes, glioma patients were well distinguished into two subgroups with survival and tumor microenvironment infiltration differences. Next, we further screened the independent prognostic genes (CTTNBP2, KIF18A, NAV1, SLAIN2, SRCIN1, TRIO, and TTBK2) using 36 prognostic-related differentially expressed MPERGs to construct a signature with risk stratification and prognostic prediction ability. An increased risk score was related to the malignant progression of glioma. Therefore, we also designed a nomogram model containing clinical factors to facilitate the clinical use of the risk signature. The prediction accuracy of the signature and nomogram model was verified using The Cancer Genome Atlas and Chinese Glioma Genome Atlas datasets. Finally, we examined the connection between the signature and tumor microenvironment. The signature positively correlated with tumor microenvironment infiltration, especially immunoinhibitors and the tumor mutation load, and negatively correlated with microsatellite instability and cancer stemness. More importantly, immune checkpoint blockade treatment and drug sensitivity analyses confirmed that this prognostic signature was helpful in anticipating the effect of immunotherapy and chemotherapy. In conclusion, this research is the first study to define and validate an MPERG-based signature closely associated with the tumor microenvironment as a reliable and independent prognostic biomarker to guide personalized choices of immunotherapy and chemotherapy for glioma patients.
Collapse
Affiliation(s)
- Wenwen Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Weilong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Lifang Pan
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310006, China
| | - Lingjie Li
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310006, China
| | - Yasi Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yuqing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaochen Zhang
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310006, China
- Correspondence: (X.Z.); (S.Z.); Tel./Fax: +86-571-5600-7650 (S.Z.)
| | - Shirong Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Correspondence: (X.Z.); (S.Z.); Tel./Fax: +86-571-5600-7650 (S.Z.)
| |
Collapse
|
10
|
Kounoupa Z, Tivodar S, Theodorakis K, Kyriakis D, Denaxa M, Karagogeos D. Rac1 and Rac3 GTPases and TPC2 are required for axonal outgrowth and migration of cortical interneurons. J Cell Sci 2023; 136:286920. [PMID: 36744839 DOI: 10.1242/jcs.260373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 01/31/2023] [Indexed: 02/07/2023] Open
Abstract
Rho GTPases, among them Rac1 and Rac3, are major transducers of extracellular signals and are involved in multiple cellular processes. In cortical interneurons, the neurons that control the balance between excitation and inhibition of cortical circuits, Rac1 and Rac3 are essential for their development. Ablation of both leads to a severe reduction in the numbers of mature interneurons found in the murine cortex, which is partially due to abnormal cell cycle progression of interneuron precursors and defective formation of growth cones in young neurons. Here, we present new evidence that upon Rac1 and Rac3 ablation, centrosome, Golgi complex and lysosome positioning is significantly perturbed, thus affecting both interneuron migration and axon growth. Moreover, for the first time, we provide evidence of altered expression and localization of the two-pore channel 2 (TPC2) voltage-gated ion channel that mediates Ca2+ release. Pharmacological inhibition of TPC2 negatively affected axonal growth and migration of interneurons. Our data, taken together, suggest that TPC2 contributes to the severe phenotype in axon growth initiation, extension and interneuron migration in the absence of Rac1 and Rac3.
Collapse
Affiliation(s)
- Zouzana Kounoupa
- Institute of Molecular Biology and Biotechnology (IMBB, FORTH), Heraklion 71110, Greece.,Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion 71110, Greece
| | - Simona Tivodar
- Institute of Molecular Biology and Biotechnology (IMBB, FORTH), Heraklion 71110, Greece.,Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion 71110, Greece
| | - Kostas Theodorakis
- Institute of Molecular Biology and Biotechnology (IMBB, FORTH), Heraklion 71110, Greece.,Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion 71110, Greece
| | - Dimitrios Kyriakis
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Myrto Denaxa
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre 'Al. Fleming', Vari, 16672, Greece
| | - Domna Karagogeos
- Institute of Molecular Biology and Biotechnology (IMBB, FORTH), Heraklion 71110, Greece.,Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion 71110, Greece
| |
Collapse
|
11
|
Wu SK, Ariffin J, Tay SC, Picone R. The variant senescence-associated secretory phenotype induced by centrosome amplification constitutes a pathway that activates hypoxia-inducible factor-1α. Aging Cell 2023; 22:e13766. [PMID: 36660875 PMCID: PMC10014068 DOI: 10.1111/acel.13766] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 01/21/2023] Open
Abstract
The senescence-associated secretory phenotype (SASP) can promote paracrine invasion while suppressing tumour growth, thus generating complex phenotypic outcomes. Likewise, centrosome amplification can induce proliferation arrest yet also facilitate tumour invasion. However, the eventual fate of cells with centrosome amplification remains elusive. Here, we report that centrosome amplification induces a variant of SASP, which constitutes a pathway activating paracrine invasion. The centrosome amplification-induced SASP is non-canonical as it lacks the archetypal detectable DNA damage and prominent NF-κB activation, but involves Rac activation and production of reactive oxygen species. Consequently, it induces hypoxia-inducible factor 1α and associated genes, including pro-migratory factors such as ANGPTL4. Of note, cellular senescence can either induce tumourigenesis through paracrine signalling or conversely suppress tumourigenesis through p53 induction. By analogy, centrosome amplification-induced SASP may therefore be one reason why extra centrosomes promote malignancy in some experimental models but are neutral in others.
Collapse
Affiliation(s)
- Selwin K. Wu
- Department of Cell BiologyHarvard Medical SchoolMassachusettsBostonUSA
- Department of Pediatric OncologyDana‐Farber Cancer InstituteMassachusettsBostonUSA
| | - Juliana Ariffin
- Department of SurgeryCancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical SchoolMassachusettsBostonUSA
- Present address:
Mechanobiology Institute & Department of Biological SciencesNational University of SingaporeSingapore
| | - Shu Chian Tay
- Mechanobiology InstituteNational University of SingaporeSingapore
| | - Remigio Picone
- Department of Cell BiologyHarvard Medical SchoolMassachusettsBostonUSA
- Department of Pediatric OncologyDana‐Farber Cancer InstituteMassachusettsBostonUSA
- Present address:
Mechanobiology Institute & Department of Biological SciencesNational University of SingaporeSingapore
| |
Collapse
|
12
|
Legátová A, Pelantová M, Rösel D, Brábek J, Škarková A. The emerging role of microtubules in invasion plasticity. Front Oncol 2023; 13:1118171. [PMID: 36860323 PMCID: PMC9969133 DOI: 10.3389/fonc.2023.1118171] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
The ability of cells to switch between different invasive modes during metastasis, also known as invasion plasticity, is an important characteristic of tumor cells that makes them able to resist treatment targeted to a particular invasion mode. Due to the rapid changes in cell morphology during the transition between mesenchymal and amoeboid invasion, it is evident that this process requires remodeling of the cytoskeleton. Although the role of the actin cytoskeleton in cell invasion and plasticity is already quite well described, the contribution of microtubules is not yet fully clarified. It is not easy to infer whether destabilization of microtubules leads to higher invasiveness or the opposite since the complex microtubular network acts differently in diverse invasive modes. While mesenchymal migration typically requires microtubules at the leading edge of migrating cells to stabilize protrusions and form adhesive structures, amoeboid invasion is possible even in the absence of long, stable microtubules, albeit there are also cases of amoeboid cells where microtubules contribute to effective migration. Moreover, complex crosstalk of microtubules with other cytoskeletal networks participates in invasion regulation. Altogether, microtubules play an important role in tumor cell plasticity and can be therefore targeted to affect not only cell proliferation but also invasive properties of migrating cells.
Collapse
Affiliation(s)
- Anna Legátová
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Markéta Pelantová
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Jan Brábek
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Aneta Škarková
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia,*Correspondence: Aneta Škarková,
| |
Collapse
|
13
|
Bonnet M, Roche F, Fagotto-Kaufmann C, Gazdagh G, Truong I, Comunale F, Barbosa S, Bonhomme M, Nafati N, Hunt D, Rodriguez MP, Chaudhry A, Shears D, Madruga M, Vansenne F, Curie A, Kajava AV, Baralle D, Fassier C, Debant A, Schmidt S. Pathogenic TRIO variants associated with neurodevelopmental disorders perturb the molecular regulation of TRIO and axon pathfinding in vivo. Mol Psychiatry 2023; 28:1527-1544. [PMID: 36717740 DOI: 10.1038/s41380-023-01963-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/26/2022] [Accepted: 01/13/2023] [Indexed: 01/31/2023]
Abstract
The RhoGEF TRIO is known to play a major role in neuronal development by controlling actin cytoskeleton remodeling, primarily through the activation of the RAC1 GTPase. Numerous de novo mutations in the TRIO gene have been identified in individuals with neurodevelopmental disorders (NDDs). We have previously established the first phenotype/genotype correlation in TRIO-associated diseases, with striking correlation between the clinical features of the individuals and the opposite modulation of RAC1 activity by TRIO variants targeting different domains. The mutations hyperactivating RAC1 are of particular interest, as they are recurrently found in patients and are associated with a severe form of NDD and macrocephaly, indicating their importance in the etiology of the disease. Yet, it remains unknown how these pathogenic TRIO variants disrupt TRIO activity at a molecular level and how they affect neurodevelopmental processes such as axon outgrowth or guidance. Here we report an additional cohort of individuals carrying a pathogenic TRIO variant that reinforces our initial phenotype/genotype correlation. More importantly, by performing conformation predictions coupled to biochemical validation, we propose a model whereby TRIO is inhibited by an intramolecular fold and NDD-associated variants relieve this inhibition, leading to RAC1 hyperactivation. Moreover, we show that in cultured primary neurons and in the zebrafish developmental model, these gain-of-function variants differentially affect axon outgrowth and branching in vitro and in vivo, as compared to loss-of-function TRIO variants. In summary, by combining clinical, molecular, cellular and in vivo data, we provide compelling new evidence for the pathogenicity of novel genetic variants targeting the TRIO gene in NDDs. We report a novel mechanism whereby the fine-tuned regulation of TRIO activity is critical for proper neuronal development and is disrupted by pathogenic mutations.
Collapse
Affiliation(s)
- Maxime Bonnet
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France
| | - Fiona Roche
- Institut de la Vision, Sorbonne University, CNRS, INSERM, Paris, France
| | - Christine Fagotto-Kaufmann
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France
| | - Gabriella Gazdagh
- Faculty of Medicine, University of Southampton, Southampton, SO16 5YA, UK.,Wessex Clinical Genetics Service, University Hospital Southampton National Health Service Foundation Trust, Southampton, SO16 5YA, UK
| | - Iona Truong
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France.,Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Franck Comunale
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France
| | - Sonia Barbosa
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France
| | - Marion Bonhomme
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France
| | - Nicolas Nafati
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, 34293, Montpellier, France
| | - David Hunt
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, SO16 5YA, UK
| | | | - Ayeshah Chaudhry
- Department of Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Deborah Shears
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Marcos Madruga
- Hospital Viamed Santa Ángela De la Cruz, Sevilla, 41014, Spain
| | - Fleur Vansenne
- Department of Clinical Genetics, University Medical Center, Groningen, 9713 GZ, Groningen, The Netherlands
| | - Aurore Curie
- Reference Center for Intellectual Disability from rare causes, Department of Child Neurology, Woman Mother and Child Hospital, Hospices Civils de Lyon, Lyon Neuroscience Research Centre, CNRS UMR5292, INSERM U1028, Université de Lyon, Bron, France
| | - Andrey V Kajava
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France
| | - Diana Baralle
- Faculty of Medicine, University of Southampton, Southampton, SO16 5YA, UK
| | - Coralie Fassier
- Institut de la Vision, Sorbonne University, CNRS, INSERM, Paris, France
| | - Anne Debant
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France.
| | - Susanne Schmidt
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
14
|
Powers RM, Hevner RF, Halpain S. The Neuron Navigators: Structure, function, and evolutionary history. Front Mol Neurosci 2023; 15:1099554. [PMID: 36710926 PMCID: PMC9877351 DOI: 10.3389/fnmol.2022.1099554] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Neuron navigators (Navigators) are cytoskeletal-associated proteins important for neuron migration, neurite growth, and axon guidance, but they also function more widely in other tissues. Recent studies have revealed novel cellular functions of Navigators such as macropinocytosis, and have implicated Navigators in human disorders of axon growth. Navigators are present in most or all bilaterian animals: vertebrates have three Navigators (NAV1-3), Drosophila has one (Sickie), and Caenorhabditis elegans has one (Unc-53). Structurally, Navigators have conserved N- and C-terminal regions each containing specific domains. The N-terminal region contains a calponin homology (CH) domain and one or more SxIP motifs, thought to interact with the actin cytoskeleton and mediate localization to microtubule plus-end binding proteins, respectively. The C-terminal region contains two coiled-coil domains, followed by a AAA+ family nucleoside triphosphatase domain of unknown activity. The Navigators appear to have evolved by fusion of N- and C-terminal region homologs present in simpler organisms. Overall, Navigators participate in the cytoskeletal response to extracellular cues via microtubules and actin filaments, in conjunction with membrane trafficking. We propose that uptake of fluid-phase cues and nutrients and/or downregulation of cell surface receptors could represent general mechanisms that explain Navigator functions. Future studies developing new models, such as conditional knockout mice or human cerebral organoids may reveal new insights into Navigator function. Importantly, further biochemical studies are needed to define the activities of the Navigator AAA+ domain, and to study potential interactions among different Navigators and their binding partners.
Collapse
Affiliation(s)
- Regina M. Powers
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Robert F. Hevner
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,Department of Pathology, UC San Diego School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Shelley Halpain
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,*Correspondence: Shelley Halpain, ✉
| |
Collapse
|
15
|
Powers RM, Daza R, Koehler AE, Courchet J, Calabrese B, Hevner RF, Halpain S. Growth cone macropinocytosis of neurotrophin receptor and neuritogenesis are regulated by neuron navigator 1. Mol Biol Cell 2022; 33:ar64. [PMID: 35352947 PMCID: PMC9561856 DOI: 10.1091/mbc.e21-12-0623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neuron navigator 1 (Nav1) is a cytoskeleton-associated protein expressed during brain development that is necessary for proper neuritogenesis, but the underlying mechanisms are poorly understood. Here we show that Nav1 is present in elongating axon tracts during mouse brain embryogenesis. We found that depletion of Nav1 in cultured neurons disrupts growth cone morphology and neurotrophin-stimulated neuritogenesis. In addition to regulating both F-actin and microtubule properties, Nav1 promotes actin-rich membrane ruffles in the growth cone and promotes macropinocytosis at those membrane ruffles, including internalization of the TrkB receptor for the neurotrophin brain-derived neurotropic factor (BDNF). Growth cone macropinocytosis is important for downstream signaling, neurite targeting, and membrane recycling, implicating Nav1 in one or more of these processes. Depletion of Nav1 also induces transient membrane blebbing via disruption of signaling in the Rho GTPase signaling pathway, supporting the novel role of Nav1 in dynamic actin-based membrane regulation at the cell periphery. These data demonstrate that Nav1 works at the interface of microtubules, actin, and plasma membrane to organize the cell periphery and promote uptake of growth and guidance cues to facilitate neural morphogenesis during development.
Collapse
Affiliation(s)
- Regina M. Powers
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037
| | - Ray Daza
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037,Department of Pathology, University of California, San Diego, La Jolla, CA 92161
| | - Alanna E. Koehler
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037,Department of Pathology, University of California, San Diego, La Jolla, CA 92161
| | - Julien Courchet
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon I, 69008 Lyon Cedex, France
| | - Barbara Calabrese
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037
| | - Robert F. Hevner
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037,Department of Pathology, University of California, San Diego, La Jolla, CA 92161
| | - Shelley Halpain
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037,*Address correspondence to: Shelley Halpain ()
| |
Collapse
|
16
|
Dietary Restriction and Rapamycin Affect Brain Aging in Mice by Attenuating Age-Related DNA Methylation Changes. Genes (Basel) 2022; 13:genes13040699. [PMID: 35456505 PMCID: PMC9030181 DOI: 10.3390/genes13040699] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/02/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023] Open
Abstract
The fact that dietary restriction (DR) and long-term rapamycin treatment (RALL) can ameliorate the aging process has been reported by many researchers. As the interface between external and genetic factors, epigenetic modification such as DNA methylation may have latent effects on the aging rate at the molecular level. To understand the mechanism behind the impacts of dietary restriction and rapamycin on aging, DNA methylation and gene expression changes were measured in the hippocampi of different-aged mice. Examining the single-base resolution of DNA methylation, we discovered that both dietary restriction and rapamycin treatment can maintain DNA methylation in a younger state compared to normal-aged mice. Through functional enrichment analysis of genes in which DNA methylation or gene expression can be affected by DR/RALL, we found that DR/RALL may retard aging through a relationship in which DNA methylation and gene expression work together not only in the same gene but also in the same biological process. This study is instructive for understanding the maintenance of DNA methylation by DR/RALL in the aging process, as well as the role of DR and RALL in the amelioration of aging.
Collapse
|
17
|
Duman JG, Blanco FA, Cronkite CA, Ru Q, Erikson KC, Mulherkar S, Saifullah AB, Firozi K, Tolias KF. Rac-maninoff and Rho-vel: The symphony of Rho-GTPase signaling at excitatory synapses. Small GTPases 2022; 13:14-47. [PMID: 33955328 PMCID: PMC9707551 DOI: 10.1080/21541248.2021.1885264] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 01/15/2023] Open
Abstract
Synaptic connections between neurons are essential for every facet of human cognition and are thus regulated with extreme precision. Rho-family GTPases, molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state, comprise a critical feature of synaptic regulation. Rho-GTPases are exquisitely controlled by an extensive suite of activators (GEFs) and inhibitors (GAPs and GDIs) and interact with many different signalling pathways to fulfill their roles in orchestrating the development, maintenance, and plasticity of excitatory synapses of the central nervous system. Among the mechanisms that control Rho-GTPase activity and signalling are cell surface receptors, GEF/GAP complexes that tightly regulate single Rho-GTPase dynamics, GEF/GAP and GEF/GEF functional complexes that coordinate multiple Rho-family GTPase activities, effector positive feedback loops, and mutual antagonism of opposing Rho-GTPase pathways. These complex regulatory mechanisms are employed by the cells of the nervous system in almost every step of development, and prominently figure into the processes of synaptic plasticity that underlie learning and memory. Finally, misregulation of Rho-GTPases plays critical roles in responses to neuronal injury, such as traumatic brain injury and neuropathic pain, and in neurodevelopmental and neurodegenerative disorders, including intellectual disability, autism spectrum disorder, schizophrenia, and Alzheimer's Disease. Thus, decoding the mechanisms of Rho-GTPase regulation and function at excitatory synapses has great potential for combatting many of the biggest current challenges in mental health.
Collapse
Affiliation(s)
- Joseph G. Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Francisco A. Blanco
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. Cronkite
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kelly C. Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ali Bin Saifullah
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
18
|
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: 1.7] [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
|
19
|
Masner M, Lujea N, Bisbal M, Acosta C, Kunda P. Linoleic and oleic acids enhance cell migration by altering the dynamics of microtubules and the remodeling of the actin cytoskeleton at the leading edge. Sci Rep 2021; 11:14984. [PMID: 34294745 PMCID: PMC8298526 DOI: 10.1038/s41598-021-94399-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/30/2021] [Indexed: 01/22/2023] Open
Abstract
Fatty acids (FA) have a multitude of biological actions on living cells. A target of their action is cell motility, a process of critical importance during cancer cell dissemination. Here, we studied the effect of unsaturated FA on ovarian cancer cell migration in vitro and its role in regulating cytoskeleton structures that are essential for cell motility. Scratch wound assays on human ovary cancer SKOV-3 cell monolayers revealed that low doses (16 μM) of linoleic acid (LA, 18:2 ω6) and oleic acid (OA; 18:1 ω9) promoted migration, while α-linolenic acid (ALA, 18:3 ω3), showed a migration rate similar to that of the control group. Single cell tracking demonstrated that LA and OA-treated cells migrated faster and were more orientated towards the wound closure than control. In vitro addition of those FA resulted in an increased number, length and protrusion speed of filopodia and also in a prominent and dynamic lamellipodia at the cell leading edge. Using time-lapse video-microscopy and FRAP we observed an increase in both the speed and frequency of actin waves associated with more mobile actin and augmented Rac1 activity. We also observed that FA induced microtubule-organizing center (MTOC)-orientation towards the cell front and affected the dynamics of microtubules (MT) in the direction of cell migration. We propose that environmental cues such as OA and LA present in ascitic fluid, should be taken into account as key factors for the regulation of cell migration.
Collapse
Affiliation(s)
- M Masner
- Centro de Investigación en Medicina Traslacional "Severo Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Naciones Unidas 420, Córdoba, Argentina
| | - N Lujea
- Centro de Investigación en Medicina Traslacional "Severo Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Naciones Unidas 420, Córdoba, Argentina
| | - M Bisbal
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - C Acosta
- Instituto de Histología y Embriología de Mendoza (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Patricia Kunda
- Centro de Investigación en Medicina Traslacional "Severo Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Naciones Unidas 420, Córdoba, Argentina.
| |
Collapse
|
20
|
Efimova N, Yang C, Chia JX, Li N, Lengner CJ, Neufeld KL, Svitkina TM. Branched actin networks are assembled on microtubules by adenomatous polyposis coli for targeted membrane protrusion. J Cell Biol 2021; 219:151902. [PMID: 32597939 PMCID: PMC7480092 DOI: 10.1083/jcb.202003091] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/26/2022] Open
Abstract
Cell migration is driven by pushing and pulling activities of the actin cytoskeleton, but migration directionality is largely controlled by microtubules. This function of microtubules is especially critical for neuron navigation. However, the underlying mechanisms are poorly understood. Here we show that branched actin filament networks, the main pushing machinery in cells, grow directly from microtubule tips toward the leading edge in growth cones of hippocampal neurons. Adenomatous polyposis coli (APC), a protein with both tumor suppressor and cytoskeletal functions, concentrates at the microtubule-branched network interface, whereas APC knockdown nearly eliminates branched actin in growth cones and prevents growth cone recovery after repellent-induced collapse. Conversely, encounters of dynamic APC-positive microtubule tips with the cell edge induce local actin-rich protrusions. Together, we reveal a novel mechanism of cell navigation involving APC-dependent assembly of branched actin networks on microtubule tips.
Collapse
Affiliation(s)
- Nadia Efimova
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - Changsong Yang
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - Jonathan X Chia
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - Ning Li
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Cell and Developmental Biology, Perelman School of Medicine and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kristi L Neufeld
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS
| | - Tatyana M Svitkina
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
21
|
Sánchez-Huertas C, Bonhomme M, Falco A, Fagotto-Kaufmann C, van Haren J, Jeanneteau F, Galjart N, Debant A, Boudeau J. The +TIP Navigator-1 is an actin-microtubule crosslinker that regulates axonal growth cone motility. J Cell Biol 2021; 219:151835. [PMID: 32497170 PMCID: PMC7480110 DOI: 10.1083/jcb.201905199] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 04/03/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
Microtubule (MT) plus-end tracking proteins (+TIPs) are central players in the coordination between the MT and actin cytoskeletons in growth cones (GCs) during axon guidance. The +TIP Navigator-1 (NAV1) is expressed in the developing nervous system, yet its neuronal functions remain poorly elucidated. Here, we report that NAV1 controls the dynamics and motility of the axonal GCs of cortical neurons in an EB1-dependent manner and is required for axon turning toward a gradient of netrin-1. NAV1 accumulates in F-actin-rich domains of GCs and binds actin filaments in vitro. NAV1 can also bind MTs independently of EB1 in vitro and crosslinks nonpolymerizing MT plus ends to actin filaments in axonal GCs, preventing MT depolymerization in F-actin-rich areas. Together, our findings pinpoint NAV1 as a key player in the actin-MT crosstalk that promotes MT persistence at the GC periphery and regulates GC steering. Additionally, we present data assigning to NAV1 an important role in the radial migration of cortical projection neurons in vivo.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Marion Bonhomme
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Amandine Falco
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Christine Fagotto-Kaufmann
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Jeffrey van Haren
- Department of Cell Biology and Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Niels Galjart
- Department of Cell Biology and Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne Debant
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Jérôme Boudeau
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| |
Collapse
|
22
|
Bircher JE, Koleske AJ. Trio family proteins as regulators of cell migration and morphogenesis in development and disease - mechanisms and cellular contexts. J Cell Sci 2021; 134:jcs248393. [PMID: 33568469 PMCID: PMC7888718 DOI: 10.1242/jcs.248393] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The well-studied members of the Trio family of proteins are Trio and kalirin in vertebrates, UNC-73 in Caenorhabditis elegans and Trio in Drosophila Trio proteins are key regulators of cell morphogenesis and migration, tissue organization, and secretion and protein trafficking in many biological contexts. Recent discoveries have linked Trio and kalirin to human disease, including neurological disorders and cancer. The genes for Trio family proteins encode a series of large multidomain proteins with up to three catalytic activities and multiple scaffolding and protein-protein interaction domains. As such, Trio family proteins engage a wide array of cell surface receptors, substrates and interaction partners to coordinate changes in cytoskeletal regulatory and protein trafficking pathways. We provide a comprehensive review of the specific mechanisms by which Trio family proteins carry out their functions in cells, highlight the biological and cellular contexts in which they occur, and relate how alterations in these functions contribute to human disease.
Collapse
Affiliation(s)
- Josie E Bircher
- Department of Molecular Biochemistry and Biophysics, Yale School of Medicine, Yale University, New Haven, CT 06511 USA
| | - Anthony J Koleske
- Department of Molecular Biochemistry and Biophysics, Yale School of Medicine, Yale University, New Haven, CT 06511 USA
| |
Collapse
|
23
|
Fujishima K, Kurisu J, Yamada M, Kengaku M. βIII spectrin controls the planarity of Purkinje cell dendrites by modulating perpendicular axon-dendrite interactions. Development 2020; 147:226102. [PMID: 33234719 DOI: 10.1242/dev.194530] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/04/2020] [Indexed: 01/14/2023]
Abstract
The mechanism underlying the geometrical patterning of axon and dendrite wiring remains elusive, despite its crucial importance in the formation of functional neural circuits. The cerebellar Purkinje cell (PC) arborizes a typical planar dendrite, which forms an orthogonal network with granule cell (GC) axons. By using electrospun nanofiber substrates, we reproduce the perpendicular contacts between PC dendrites and GC axons in culture. In the model system, PC dendrites show a preference to grow perpendicularly to aligned GC axons, which presumably contribute to the planar dendrite arborization in vivo We show that βIII spectrin, a causal protein for spinocerebellar ataxia type 5, is required for the biased growth of dendrites. βIII spectrin deficiency causes actin mislocalization and excessive microtubule invasion in dendritic protrusions, resulting in abnormally oriented branch formation. Furthermore, disease-associated mutations affect the ability of βIII spectrin to control dendrite orientation. These data indicate that βIII spectrin organizes the mouse dendritic cytoskeleton and thereby regulates the oriented growth of dendrites with respect to the afferent axons.
Collapse
Affiliation(s)
- Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Junko Kurisu
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Midori Yamada
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Mineko Kengaku
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
24
|
Katrancha SM, Shaw JE, Zhao AY, Myers SA, Cocco AR, Jeng AT, Zhu M, Pittenger C, Greer CA, Carr SA, Xiao X, Koleske AJ. Trio Haploinsufficiency Causes Neurodevelopmental Disease-Associated Deficits. Cell Rep 2020; 26:2805-2817.e9. [PMID: 30840899 PMCID: PMC6436967 DOI: 10.1016/j.celrep.2019.02.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/22/2018] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Heterozygous coding mutations in TRIO are associated with neurodevelopmental disorders, including autism, schizophrenia, bipolar disorder, and epilepsy, and impair TRIO's biochemical activities. To model mutant alleles, we ablated one or both Trio alleles from excitatory neurons in the cortex and hippocampus of mice. Trio haploinsufficiency increases anxiety and impairs social preference and motor coordination. Trio loss reduces forebrain size and dendritic arborization but increases dendritic spine densities. Cortical synapses in Trio haploinsufficient mice are small, exhibit pre- and postsynaptic deficits, and cannot undergo long-term potentiation. Similar phenotypes are observed in Trio knockout mice. Overall, Trio haploinsufficiency causes severe disease-relevant deficits in behavior and neuronal structure and function. Interestingly, phosphodiesterase 4A5 (PDE4A5) levels are reduced and protein kinase A (PKA) signaling is increased when TRIO levels are reduced. Elevation of PDE4A5 and drug-based attenuation of PKA signaling rescue Trio haploinsufficiency-related dendritic spine defects, suggesting an avenue for therapeutic intervention for TRIO-related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sara Marie Katrancha
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA
| | - Juliana E Shaw
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Amy Y Zhao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA
| | - Samuel A Myers
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Amanda T Jeng
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA
| | - Minsheng Zhu
- Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Christopher Pittenger
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Psychiatry, Yale University, New Haven, CT 06510, USA; Child Study Center, Yale University, New Haven, CT 06510, USA
| | - Charles A Greer
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA; Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Steven A Carr
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiao Xiao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai 200433, China.
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
25
|
Abdelkarim H, Hitchinson B, Qu X, Banerjee A, Komarova YA, Gaponenko V. NMR resonance assignment and structure prediction of the C-terminal domain of the microtubule end-binding protein 3. PLoS One 2020; 15:e0232338. [PMID: 32421702 PMCID: PMC7233555 DOI: 10.1371/journal.pone.0232338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 04/13/2020] [Indexed: 11/29/2022] Open
Abstract
End-binding proteins (EBs) associate with the growing microtubule plus ends to regulate microtubule dynamics as well as the interaction with intracellular structures. EB3 contributes to pathological vascular leakage through interacting with the inositol 1,4,5-trisphosphate receptor 3 (IP3R3), a calcium channel located at the endoplasmic reticulum membrane. The C-terminal domain of EB3 (residues 200–281) is functionally important for this interaction because it contains the effector binding sites, a prerequisite for EB3 activity and specificity. Structural data for this domain is limited. Here, we report the backbone chemical shift assignments for the human EB3 C-terminal domain and computationally explore its EB3 conformations. Backbone assignments, along with computational models, will allow future investigation of EB3 structural dynamics, interactions with effectors, and will facilitate the development of novel EB3 inhibitors.
Collapse
Affiliation(s)
- Hazem Abdelkarim
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Ben Hitchinson
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Xinyan Qu
- Department of Pharmacology and the Center for Lung Biology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Avik Banerjee
- Department of Chemistry, University of Illinois, Chicago, IL, United States of America
| | - Yulia A. Komarova
- Department of Pharmacology and the Center for Lung Biology, University of Illinois at Chicago, Chicago, IL, United States of America
- * E-mail: (YAK); (VG)
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
- * E-mail: (YAK); (VG)
| |
Collapse
|
26
|
Abstract
Directed cell migration is critical for embryogenesis and organ development, wound healing and the immune response. Microtubules are dynamic polymers that control directional migration through a number of coordinated processes: microtubules are the tracks for long-distance intracellular transport, crucial for delivery of new membrane components and signalling molecules to the leading edge of a migrating cell and the recycling of adhesion receptors. Microtubules act as force generators and compressive elements to support sustained cell protrusions. The assembly and disassembly of microtubules is coupled to Rho GTPase signalling, thereby controlling actin polymerisation, myosin-driven contractility and the turnover of cellular adhesions locally. Cross-talk of actin and microtubule dynamics is mediated through a number of common binding proteins and regulators. Furthermore, cortical microtubule capture sites are physically linked to focal adhesions, facilitating the delivery of secretory vesicles and efficient cross-talk. Here we summarise the diverse functions of microtubules during cell migration, aiming to show how they contribute to the spatially and temporally coordinated sequence of events that permit efficient, directional and persistent migration.
Collapse
|
27
|
Arbeev KG, Bagley O, Ukraintseva SV, Wu D, Duan H, Kulminski AM, Stallard E, Christensen K, Lee JH, Thyagarajan B, Zmuda JM, Yashin AI. Genetics of physiological dysregulation: findings from the long life family study using joint models. Aging (Albany NY) 2020; 12:5920-5947. [PMID: 32235003 PMCID: PMC7185144 DOI: 10.18632/aging.102987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
Recently, Mahalanobis distance (DM) was suggested as a statistical measure of physiological dysregulation in aging individuals. We constructed DM variants using sets of biomarkers collected at the two visits of the Long Life Family Study (LLFS) and performed joint analyses of longitudinal observations of DM and follow-up mortality in LLFS using joint models. We found that DM is significantly associated with mortality (hazard ratio per standard deviation: 1.31 [1.16, 1.48] to 2.22 [1.84, 2.67]) after controlling for age and other covariates. GWAS of random intercepts and slopes of DM estimated from joint models found a genome-wide significant SNP (rs12652543, p=7.2×10-9) in the TRIO gene associated with the slope of DM constructed from biomarkers declining in late life. Review of biological effects of genes corresponding to top SNPs from GWAS of DM slopes revealed that these genes are broadly involved in cancer prognosis and axon guidance/synapse function. Although axon growth is mainly observed during early development, the axon guidance genes can function in adults and contribute to maintenance of neural circuits and synaptic plasticity. Our results indicate that decline in axons' ability to maintain complex regulatory networks may potentially play an important role in the increase in physiological dysregulation during aging.
Collapse
Affiliation(s)
- Konstantin G Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Svetlana V Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Deqing Wu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Hongzhe Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Kaare Christensen
- Danish Aging Research Center, Department of Public Health, University of Southern Denmark 5000, Odense C, Denmark
| | - Joseph H Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA.,G. H. Sergievsky Center, Columbia University, New York, NY 10032, USA.,Departments of Epidemiology and Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph M Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anatoliy I Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| |
Collapse
|
28
|
Barbosa S, Greville-Heygate S, Bonnet M, Godwin A, Fagotto-Kaufmann C, Kajava AV, Laouteouet D, Mawby R, Wai HA, Dingemans AJ, Hehir-Kwa J, Willems M, Capri Y, Mehta SG, Cox H, Goudie D, Vansenne F, Turnpenny P, Vincent M, Cogné B, Lesca G, Hertecant J, Rodriguez D, Keren B, Burglen L, Gérard M, Putoux A, Cantagrel V, Siquier-Pernet K, Rio M, Banka S, Sarkar A, Steeves M, Parker M, Clement E, Moutton S, Tran Mau-Them F, Piton A, de Vries BB, Guille M, Debant A, Schmidt S, Baralle D, Baralle D. Opposite Modulation of RAC1 by Mutations in TRIO Is Associated with Distinct, Domain-Specific Neurodevelopmental Disorders. Am J Hum Genet 2020; 106:338-355. [PMID: 32109419 PMCID: PMC7058823 DOI: 10.1016/j.ajhg.2020.01.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022] Open
Abstract
The Rho-guanine nucleotide exchange factor (RhoGEF) TRIO acts as a key regulator of neuronal migration, axonal outgrowth, axon guidance, and synaptogenesis by activating the GTPase RAC1 and modulating actin cytoskeleton remodeling. Pathogenic variants in TRIO are associated with neurodevelopmental diseases, including intellectual disability (ID) and autism spectrum disorders (ASD). Here, we report the largest international cohort of 24 individuals with confirmed pathogenic missense or nonsense variants in TRIO. The nonsense mutations are spread along the TRIO sequence, and affected individuals show variable neurodevelopmental phenotypes. In contrast, missense variants cluster into two mutational hotspots in the TRIO sequence, one in the seventh spectrin repeat and one in the RAC1-activating GEFD1. Although all individuals in this cohort present with developmental delay and a neuro-behavioral phenotype, individuals with a pathogenic variant in the seventh spectrin repeat have a more severe ID associated with macrocephaly than do most individuals with GEFD1 variants, who display milder ID and microcephaly. Functional studies show that the spectrin and GEFD1 variants cause a TRIO-mediated hyper- or hypo-activation of RAC1, respectively, and we observe a striking correlation between RAC1 activation levels and the head size of the affected individuals. In addition, truncations in TRIO GEFD1 in the vertebrate model X. tropicalis induce defects that are concordant with the human phenotype. This work demonstrates distinct clinical and molecular disorders clustering in the GEFD1 and seventh spectrin repeat domains and highlights the importance of tight control of TRIO-RAC1 signaling in neuronal development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Diana Baralle
- Wessex Clinical Genetics, University Hospital Southampton National Health Service Foundation Trust, Southampton SO16 5YA, UK; Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
29
|
Li D, Ding X, Xie M, Huang Z, Han P, Tian D, Xia L. CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis. Am J Cancer Res 2020; 10:3749-3766. [PMID: 32206120 PMCID: PMC7069094 DOI: 10.7150/thno.42596] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/03/2020] [Indexed: 01/13/2023] Open
Abstract
Rationale: Emerging evidence suggests that noncentrosomal microtubules play an essential role in intracellular transport, cell polarity and cell motility. Whether these noncentrosomal microtubules exist or function in cancer cells remains unclear. Methods: The expression and prognostic values of CAMSAP2 and its functional targets were analyzed by immunohistochemistry in two independent HCC cohorts. Immunofluorescence and co-immunoprecipitation were used for detection of CAMSAP2-decorated noncentrosomal microtubule. Chromatin immunoprecipitation and luciferase report assays were used to determine the c-Jun binding sites in HDAC6 promoter region. In vitro migration and invasion assays and in vivo orthotopic metastatic models were utilized to investigate invasion and metastasis. Results: We reported a microtubule minus‑end‑targeting protein, CAMSAP2, is significantly upregulated in hepatocellular carcinoma (HCC) and correlated with poor prognosis. CAMSAP2 was specifically deposited on microtubule minus ends to serve as a “seed” for noncentrosomal microtubule outgrowth in HCC cells. Upon depletion of CAMSAP2, the noncentrosomal microtubule array was transformed into a completely radial centrosomal pattern, thereby impairing HCC cell migration and invasion. We further demonstrated that CAMSAP2 cooperates with EB1 to regulate microtubule dynamics and invasive cell migration via Trio/Rac1 signaling. Strikingly, both immunofluorescence staining and western blotting showed that CAMSAP2 depletion strongly reduced the abundance of acetylated microtubules in HCC cells. Our results revealed that HDAC6, a promising target for cancer therapy, was inversely downregulated in HCC and uniquely endowed with tumor-suppressive activity by regulation CAMSAP2-mediated microtubule acetylation. Mechanistically, CAMSAP2 activates c-Jun to induce transrepression of HDAC6 through Trio-dependent Rac1/JNK pathway. Furthermore, NSC23766, a Rac1-specific inhibitor significantly inhibited CAMSAP2-mediated HCC invasion and metastasis. Conclusions: CAMSAP2 is functionally, mechanistically, and clinically oncogenic in HCC. Targeting CAMSAP2-mediated noncentrosomal microtubule acetylation may provide new therapeutic strategies for HCC metastasis.
Collapse
|
30
|
Much More Than a Scaffold: Cytoskeletal Proteins in Neurological Disorders. Cells 2020; 9:cells9020358. [PMID: 32033020 PMCID: PMC7072452 DOI: 10.3390/cells9020358] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/08/2023] Open
Abstract
Recent observations related to the structure of the cytoskeleton in neurons and novel cytoskeletal abnormalities involved in the pathophysiology of some neurological diseases are changing our view on the function of the cytoskeletal proteins in the nervous system. These efforts allow a better understanding of the molecular mechanisms underlying neurological diseases and allow us to see beyond our current knowledge for the development of new treatments. The neuronal cytoskeleton can be described as an organelle formed by the three-dimensional lattice of the three main families of filaments: actin filaments, microtubules, and neurofilaments. This organelle organizes well-defined structures within neurons (cell bodies and axons), which allow their proper development and function through life. Here, we will provide an overview of both the basic and novel concepts related to those cytoskeletal proteins, which are emerging as potential targets in the study of the pathophysiological mechanisms underlying neurological disorders.
Collapse
|
31
|
The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019; 8:cells8040362. [PMID: 31003495 PMCID: PMC6523135 DOI: 10.3390/cells8040362] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
32
|
Cheng HW, Hsiao CT, Chen YQ, Huang CM, Chan SI, Chiou A, Kuo JC. Centrosome guides spatial activation of Rac to control cell polarization and directed cell migration. Life Sci Alliance 2019; 2:2/1/e201800135. [PMID: 30737247 PMCID: PMC6369537 DOI: 10.26508/lsa.201800135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 01/01/2023] Open
Abstract
The centrosome acts as a controller by balancing the formation of centrosomal and acentrosomal microtubules, the modulation of focal adhesion signaling and the activation of local Rac1 at the cell front, which then coordinates cell polarization during directed cell migration. Directed cell migration requires centrosome-mediated cell polarization and dynamical control of focal adhesions (FAs). To examine how FAs cooperate with centrosomes for directed cell migration, we used centrosome-deficient cells and found that loss of centrosomes enhanced the formation of acentrosomal microtubules, which failed to form polarized structures in wound-edge cells. In acentrosomal cells, we detected higher levels of Rac1-guanine nucleotide exchange factor TRIO (Triple Functional Domain Protein) on microtubules and FAs. Acentrosomal microtubules deliver TRIO to FAs for Rac1 regulation. Indeed, centrosome disruption induced excessive Rac1 activation around the cell periphery via TRIO, causing rapid FA turnover, a disorganized actin meshwork, randomly protruding lamellipodia, and loss of cell polarity. This study reveals the importance of centrosomes to balance the assembly of centrosomal and acentrosomal microtubules and to deliver microtubule-associated TRIO proteins to FAs at the cell front for proper spatial activation of Rac1, FA turnover, lamillipodial protrusion, and cell polarization, thereby allowing directed cell migration.
Collapse
Affiliation(s)
- Hung-Wei Cheng
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Te Hsiao
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yin-Quan Chen
- Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan.,Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| | - Chi-Ming Huang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Seng-I Chan
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Arthur Chiou
- Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan .,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
33
|
Abstract
The cytoskeleton is crucially important for the assembly of cell-cell junctions and the homeostatic regulation of their functions. Junctional proteins act, in turn, as anchors for cytoskeletal filaments, and as regulators of cytoskeletal dynamics and signalling proteins. The cross-talk between junctions and the cytoskeleton is critical for the morphogenesis and physiology of epithelial and other tissues, but is not completely understood. Microtubules are implicated in the delivery of junctional proteins to cell-cell contact sites, in the differentiation and spatial organization of the cytoplasm, and in the stabilization of the barrier and adhesive functions of junctions. Here we focus on the relationships between microtubules and junctions of vertebrate epithelial cells. We highlight recent discoveries on the molecular underpinnings of microtubule-junction interactions, and report new data about the interaction of cingulin and paracingulin with microtubules. We also propose a possible new role of junctions as “molecular sinks” for microtubule-associated signalling proteins.
Collapse
Affiliation(s)
- Ekaterina Vasileva
- a Department of Cell Biology, Faculty of Sciences and Institute for Genetics and Genomics in Geneva (iGE3) , University of Geneva , Geneva , Switzerland
| | - Sandra Citi
- a Department of Cell Biology, Faculty of Sciences and Institute for Genetics and Genomics in Geneva (iGE3) , University of Geneva , Geneva , Switzerland
| |
Collapse
|
34
|
Roth D, Fitton BP, Chmel NP, Wasiluk N, Straube A. Spatial positioning of EB family proteins at microtubule tips involves distinct nucleotide-dependent binding properties. J Cell Sci 2018; 132:jcs.219550. [PMID: 30262468 PMCID: PMC6398475 DOI: 10.1242/jcs.219550] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/20/2018] [Indexed: 12/25/2022] Open
Abstract
EB proteins track the ends of growing microtubules and regulate microtubule dynamics both directly and by acting as the hub of the tip-tracking network. Mammalian cells express cell type-specific combinations of three EB proteins with different cellular roles. Here, we reconstitute EB1, EB2 and EB3 tip tracking in vitro. We find that all three EBs show rapid exchange at the microtubule tip and that their signal correlates to the microtubule assembly rate. However, the three signals differ in their maxima and position from the microtubule tip. Using microtubules built with nucleotide analogues and site-directed mutagenesis, we show that EB2 prefers binding to microtubule lattices containing a 1:1 mixture of different nucleotides and its distinct binding specificity is conferred by amino acid substitutions at the right-hand-side interface of the EB microtubule-binding domain with tubulin. Our data are consistent with the model that all three EB paralogues sense the nucleotide state of both β-tubulins flanking their binding site. Their different profile of preferred binding sites contributes to occupying spatially distinct domains at the temporally evolving microtubule tip structure. Summary:In vitro reconstitution of tip tracking with EB1, EB2 and EB3 shows that these three proteins sense the nucleotide state of both β-tubulins flanking their binding site.
Collapse
Affiliation(s)
- Daniel Roth
- Centre for Mechanochemical Cell Biology (CMCB), University of Warwick, Coventry CV4 7AL, UK.,Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Benjamin P Fitton
- Centre for Mechanochemical Cell Biology (CMCB), University of Warwick, Coventry CV4 7AL, UK.,Molecular Organisation and Assembly in Cells (MOAC) Doctoral Training Centre, University of Warwick, Coventry CV4 7AL, UK
| | - Nikola P Chmel
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | - Natalia Wasiluk
- Centre for Mechanochemical Cell Biology (CMCB), University of Warwick, Coventry CV4 7AL, UK
| | - Anne Straube
- Centre for Mechanochemical Cell Biology (CMCB), University of Warwick, Coventry CV4 7AL, UK .,Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
35
|
Nørgaard S, Deng S, Cao W, Pocock R. Distinct CED-10/Rac1 domains confer context-specific functions in development. PLoS Genet 2018; 14:e1007670. [PMID: 30265669 PMCID: PMC6179291 DOI: 10.1371/journal.pgen.1007670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/10/2018] [Accepted: 08/30/2018] [Indexed: 11/18/2022] Open
Abstract
Rac GTPases act as master switches to coordinate multiple interweaved signaling pathways. A major function for Rac GTPases is to control neurite development by influencing downstream effector molecules and pathways. In Caenorhabditis elegans, the Rac proteins CED-10, RAC-2 and MIG-2 act in parallel to control axon outgrowth and guidance. Here, we have identified a single glycine residue in the CED-10/Rac1 Switch 1 region that confers a non-redundant function in axon outgrowth but not guidance. Mutation of this glycine to glutamic acid (G30E) reduces GTP binding and inhibits axon outgrowth but does not affect other canonical CED-10 functions. This demonstrates previously unappreciated domain-specific functions within the CED-10 protein. Further, we reveal that when CED-10 function is diminished, the adaptor protein NAB-1 (Neurabin) and its interacting partner SYD-1 (Rho-GAP-like protein) can act as inhibitors of axon outgrowth. Together, we reveal that specific domains and residues within Rac GTPases can confer context-dependent functions during animal development. Brain development requires that neurite outgrowth and guidance are precisely regulated. Previous studies have shown that molecular switch proteins called Rac GTPases perform redundant functions in controlling neurite development. Using a pair of bilateral neurons in the nematode Caenorhabditis elegans to model neurite development, we found that a single amino acid in a conserved domain of the Rac GTPase CED-10 is crucial for controlling neurite outgrowth in a partially non-redundant manner. Further, we revealed that lesions in discrete domains in the CED-10 protein lead to distinct developmental defects. Therefore, our in vivo study proposes that regulation of distinct signalling pathways through Rac GTPase protein domains can drive different developmental outcomes.
Collapse
Affiliation(s)
- Steffen Nørgaard
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Shuer Deng
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Wei Cao
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
36
|
Masschaele D, Wauman J, Vandemoortele G, De Sutter D, De Ceuninck L, Eyckerman S, Tavernier J. High-Confidence Interactome for RNF41 Built on Multiple Orthogonal Assays. J Proteome Res 2018; 17:1348-1360. [PMID: 29560723 DOI: 10.1021/acs.jproteome.7b00704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Ring finger protein 41 (RNF41) is an E3 ubiquitin ligase involved in the ubiquitination and degradation of many proteins including ErbB3 receptors, BIRC6, and parkin. Next to this, RNF41 regulates the intracellular trafficking of certain JAK2-associated cytokine receptors by ubiquitinating and suppressing USP8, which, in turn, destabilizes the ESCRT-0 complex. To further elucidate the function of RNF41 we used different orthogonal approaches to reveal the RNF41 protein complex: affinity purification-mass spectrometry, BioID, and Virotrap. We combined these results with known data sets for RNF41 obtained with microarray MAPPIT and Y2H screens. This way, we establish a comprehensive high-resolution interactome network comprising 175 candidate protein partners. To remove potential methodological artifacts from this network, we distilled the data into a high-confidence interactome map by retaining a total of 19 protein hits identified in two or more of the orthogonal methods. AP2S1, a novel RNF41 interaction partner, was selected from this high-confidence interactome for further functional validation. We reveal a role for AP2S1 in leptin and LIF receptor signaling and show that RNF41 stabilizes and relocates AP2S1.
Collapse
Affiliation(s)
- Delphine Masschaele
- Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium.,Center for Medical Biotechnology, VIB , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium
| | - Joris Wauman
- Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium.,Center for Medical Biotechnology, VIB , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium
| | - Giel Vandemoortele
- Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium.,Center for Medical Biotechnology, VIB , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium
| | - Delphine De Sutter
- Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium.,Center for Medical Biotechnology, VIB , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium
| | - Leentje De Ceuninck
- Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium.,Center for Medical Biotechnology, VIB , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium
| | - Sven Eyckerman
- Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium.,Center for Medical Biotechnology, VIB , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium
| | - Jan Tavernier
- Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium.,Center for Medical Biotechnology, VIB , Albert Baertsoenkaai 3 , B-9000 Ghent , Belgium
| |
Collapse
|
37
|
van Haren J, Charafeddine RA, Ettinger A, Wang H, Hahn KM, Wittmann T. Local control of intracellular microtubule dynamics by EB1 photodissociation. Nat Cell Biol 2018; 20:252-261. [PMID: 29379139 PMCID: PMC5826794 DOI: 10.1038/s41556-017-0028-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 12/13/2017] [Indexed: 12/18/2022]
Abstract
End-binding proteins, EBs, are adaptors that recruit functionally diverse +TIP proteins to growing microtubule plus ends. To test with high spatial and temporal accuracy how, when and where +TIP protein complexes contribute to dynamic cell biology, we developed a photo-inactivated EB1 variant (π-EB1) by inserting a blue light-sensitive protein-protein interaction module between the microtubule- and +TIP-binding domains of EB1. π-EB1 replaces endogenous EB1 function in the absence of blue light. In contrast, blue light-mediated π-EB1 photo-dissociation results in rapid +TIP complex disassembly, and acutely and reversibly attenuates microtubule growth independent of microtubule end association of the microtubule polymerase CKAP5 (ch-TOG, XMAP215). Local π-EB1 photo-dissociation allows subcellular microtubule dynamics control at the second and micrometre scale, and elicits aversive turning of migrating cancer cells. Importantly, light-mediated domain splitting can serve as template to optically control other intracellular protein activities.
Collapse
Affiliation(s)
- Jeffrey van Haren
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Rabab A Charafeddine
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Andreas Ettinger
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA.,Institute of Epigenetics and Stem Cells, Helmholtz Center Munich, München, Germany
| | - Hui Wang
- University of North Carolina, Chapel Hill, NC, USA
| | - Klaus M Hahn
- University of North Carolina, Chapel Hill, NC, USA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA.
| |
Collapse
|
38
|
Klei LR, Hu D, Panek R, Alfano DN, Bridwell RE, Bailey KM, Oravecz-Wilson KI, Concel VJ, Hess EM, Van Beek M, Delekta PC, Gu S, Watkins SC, Ting AT, Gough PJ, Foley KP, Bertin J, McAllister-Lucas LM, Lucas PC. MALT1 Protease Activation Triggers Acute Disruption of Endothelial Barrier Integrity via CYLD Cleavage. Cell Rep 2017; 17:221-232. [PMID: 27681433 DOI: 10.1016/j.celrep.2016.08.080] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 07/14/2016] [Accepted: 08/23/2016] [Indexed: 12/26/2022] Open
Abstract
Microvascular endothelial cells maintain a tight barrier to prevent passage of plasma and circulating immune cells into the extravascular tissue compartment, yet endothelial cells respond rapidly to vasoactive substances, including thrombin, allowing transient paracellular permeability. This response is a cornerstone of acute inflammation, but the mechanisms responsible are still incompletely understood. Here, we demonstrate that thrombin triggers MALT1 to proteolytically cleave cylindromatosis (CYLD). Fragmentation of CYLD results in microtubule disruption and a cascade of events leading to endothelial cell retraction and an acute permeability response. This finding reveals an unexpected role for the MALT1 protease, which previously has been viewed mostly as a driver of pro-inflammatory NF-κB signaling in lymphocytes. Thus, MALT1 not only promotes immune cell activation but also acutely regulates endothelial cell biology, actions that together facilitate tissue inflammation. Pharmacologic inhibition of MALT1 may therefore have synergistic impact by targeting multiple disparate steps in the overall inflammatory response.
Collapse
Affiliation(s)
- Linda R Klei
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Dong Hu
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Robert Panek
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Danielle N Alfano
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Rachel E Bridwell
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Kelly M Bailey
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Vincent J Concel
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Emily M Hess
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Matthew Van Beek
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Phillip C Delekta
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Shufang Gu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Simon C Watkins
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Adrian T Ting
- Immunology Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19406, USA
| | - Kevin P Foley
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19406, USA
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19406, USA
| | - Linda M McAllister-Lucas
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Peter C Lucas
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
39
|
Lin D, Liang Y, Jing X, Chen Y, Lei M, Zeng Z, Zhou T, Wu X, Peng S, Zheng D, Huang K, Yang L, Xiao S, Liu J, Tao E. Microarray analysis of an synthetic α-synuclein induced cellular model reveals the expression profile of long non-coding RNA in Parkinson's disease. Brain Res 2017; 1678:384-396. [PMID: 29137975 DOI: 10.1016/j.brainres.2017.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 01/10/2023]
Abstract
Long non-coding RNAs (lncRNAs) are a new research focus that are reported to influence the pathogenetic process of neurodegenerative disorders. To uncover new disease-associated genes and their relevant mechanisms, we carried out a gene microarray analysis based on a Parkinson's disease (PD) in vitro model induced by α-synuclein oligomers. This cellular model induced by 25 μmol/L α-synuclein oligomers has been confirmed to show the stable, transmissible neurotoxicity of α-synuclein, a typical PD pathological marker. And several differentially expressed lncRNAs and mRNAs were identified in this model, such as G046036, G030771, AC009365.4, RPS14P3, CTB-11I22.1, and G007549. Subsequent ceRNA analysis determined the potential relationships between these lncRNAs and their associated mRNAs and microRNAs. The results of the present study widen our horizon of PD susceptibility genes and provide new pathways towards efficient diagnostic biomarkers and therapeutic targets for PD.
Collapse
Affiliation(s)
- D Lin
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - Y Liang
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - X Jing
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - Y Chen
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - M Lei
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - Z Zeng
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - T Zhou
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - X Wu
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - S Peng
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - D Zheng
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - K Huang
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - L Yang
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - S Xiao
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - J Liu
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China
| | - E Tao
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510080, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China.
| |
Collapse
|
40
|
Reeves WM, Wu Y, Harder MJ, Veeman MT. Functional and evolutionary insights from the Ciona notochord transcriptome. Development 2017; 144:3375-3387. [PMID: 28928284 DOI: 10.1242/dev.156174] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022]
Abstract
The notochord of the ascidian Ciona consists of only 40 cells, and is a longstanding model for studying organogenesis in a small, simple embryo. Here, we perform RNAseq on flow-sorted notochord cells from multiple stages to define a comprehensive Ciona notochord transcriptome. We identify 1364 genes with enriched expression and extensively validate the results by in situ hybridization. These genes are highly enriched for Gene Ontology terms related to the extracellular matrix, cell adhesion and cytoskeleton. Orthologs of 112 of the Ciona notochord genes have known notochord expression in vertebrates, more than twice as many as predicted by chance alone. This set of putative effector genes with notochord expression conserved from tunicates to vertebrates will be invaluable for testing hypotheses about notochord evolution. The full set of Ciona notochord genes provides a foundation for systems-level studies of notochord gene regulation and morphogenesis. We find only modest overlap between this set of notochord-enriched transcripts and the genes upregulated by ectopic expression of the key notochord transcription factor Brachyury, indicating that Brachyury is not a notochord master regulator gene as strictly defined.
Collapse
Affiliation(s)
- Wendy M Reeves
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Yuye Wu
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Matthew J Harder
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Michael T Veeman
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
41
|
Novozhylov DO, Karpov PA, Blume YB. Bioinformatic search for Ca2+- and calmodulin-dependent protein kinases potentially associated with the regulation of plant cytoskeleton. CYTOL GENET+ 2017. [DOI: 10.3103/s0095452717040053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Song Z, Yang W, Zhou X, Yang L, Zhao D. Lithium alleviates neurotoxic prion peptide-induced synaptic damage and neuronal death partially by the upregulation of nuclear target REST and the restoration of Wnt signaling. Neuropharmacology 2017; 123:332-348. [PMID: 28545972 DOI: 10.1016/j.neuropharm.2017.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/30/2017] [Accepted: 05/20/2017] [Indexed: 12/17/2022]
Abstract
Prion diseases are a group of infectious neurodegenerative diseases characterized by multiple neuropathological hallmarks, including accumulation of PrPSc, synaptic damage, and neuronal death. We previously reported that the repressor element 1-silencing transcription factor (REST), a novel neuroprotective marker in neurodegeneration, protects neurons against neurotoxic peptide (PrP106-126)-induced neurotoxicity, but fails to maintain survival following prolonged exposure to PrP106-126. Because Wnt signaling partially induces REST and is activated by lithium, we investigated the effects of lithium on REST in prion diseases. Lithium restores nuclear expression of REST, which is essential for regulating survival proteins. Lithium also mimics neuroprotective functions when REST is blocked, and these beneficial effects are additive with REST overexpression under physiological conditions. Reciprocally, under PrP106-126-stimulated pathological conditions, REST plays a critical role in the neuroprotective mechanisms of lithium treatment. Although lithium recovers Wnt signaling by inhibiting glycogen synthase kinase-3β and stabilizing β-catenin, restores survival associated proteins after exposure to PrP106-126 in primary cortical neurons. Knockdown of REST significantly suppresses the neuroprotective function of lithium. Conversely, overexpression of REST partially recovers its actions. Notably, lithium directly alleviates PrP106-126-induced synaptic damage and neuronal cell death by preventing changes in presynaptic and postsynaptic marker proteins and promoting survival pathways also partially via the expression of REST. Our results suggest that REST acts as a novel and important nuclear target for lithium. We hypothesize that PrP106-126-stimulated neurotoxicity induces Wnt signaling dysfunction and lithium mimics this signaling cascade, suggesting that lithium should be considered as a potential therapeutic agent against prion diseases.
Collapse
Affiliation(s)
- Zhiqi Song
- The State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Wei Yang
- The State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiangmei Zhou
- The State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Lifeng Yang
- The State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Deming Zhao
- The State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
43
|
Neuronal polarization: From spatiotemporal signaling to cytoskeletal dynamics. Mol Cell Neurosci 2017; 84:11-28. [PMID: 28363876 DOI: 10.1016/j.mcn.2017.03.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/24/2017] [Accepted: 03/26/2017] [Indexed: 12/20/2022] Open
Abstract
Neuronal polarization establishes distinct molecular structures to generate a single axon and multiple dendrites. Studies over the past years indicate that this efficient separation is brought about by a network of feedback loops. Axonal growth seems to play a major role in fueling those feedback loops and thereby stabilizing neuronal polarity. Indeed, various effectors involved in feedback loops are pivotal for axonal growth by ultimately acting on the actin and microtubule cytoskeleton. These effectors have key roles in interconnecting actin and microtubule dynamics - a mechanism crucial to commanding the growth of axons. We propose a model connecting signaling with cytoskeletal dynamics and neurite growth to better describe the underlying processes involved in neuronal polarization. We will discuss the current views on feedback loops and highlight the current limits of our understanding.
Collapse
|
44
|
Abstract
ABSTRACT
Three-dimensional (3D) cell motility underlies essential processes, such as embryonic development, tissue repair and immune surveillance, and is involved in cancer progression. Although the cytoskeleton is a well-studied regulator of cell migration, most of what we know about its functions originates from studies conducted in two-dimensional (2D) cultures. This research established that the microtubule network mediates polarized trafficking and signaling that are crucial for cell shape and movement in 2D. In parallel, developments in light microscopy and 3D cell culture systems progressively allowed to investigate cytoskeletal functions in more physiologically relevant settings. Interestingly, several studies have demonstrated that microtubule involvement in cell morphogenesis and motility can differ in 2D and 3D environments. In this Commentary, we discuss these differences and their relevance for the understanding the role of microtubules in cell migration in vivo. We also provide an overview of microtubule functions that were shown to control cell shape and motility in 3D matrices and discuss how they can be investigated further by using physiologically relevant models.
Collapse
Affiliation(s)
- Benjamin P. Bouchet
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, Utrecht 3584 CH, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, Utrecht 3584 CH, The Netherlands
| |
Collapse
|
45
|
Ionic liquid-based method for direct proteome characterization of velvet antler cartilage. Talanta 2016; 161:541-546. [DOI: 10.1016/j.talanta.2016.08.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/22/2016] [Accepted: 08/30/2016] [Indexed: 11/19/2022]
|
46
|
Terry-Lorenzo RT, Torres VI, Wagh D, Galaz J, Swanson SK, Florens L, Washburn MP, Waites CL, Gundelfinger ED, Reimer RJ, Garner CC. Trio, a Rho Family GEF, Interacts with the Presynaptic Active Zone Proteins Piccolo and Bassoon. PLoS One 2016; 11:e0167535. [PMID: 27907191 PMCID: PMC5132261 DOI: 10.1371/journal.pone.0167535] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/15/2016] [Indexed: 12/17/2022] Open
Abstract
Synaptic vesicles (SVs) fuse with the plasma membrane at a precise location called the presynaptic active zone (AZ). This fusion is coordinated by proteins embedded within a cytoskeletal matrix assembled at the AZ (CAZ). In the present study, we have identified a novel binding partner for the CAZ proteins Piccolo and Bassoon. This interacting protein, Trio, is a member of the Dbl family of guanine nucleotide exchange factors (GEFs) known to regulate the dynamic assembly of actin and growth factor dependent axon guidance and synaptic growth. Trio was found to interact with the C-terminal PBH 9/10 domains of Piccolo and Bassoon via its own N-terminal Spectrin repeats, a domain that is also critical for its localization to the CAZ. Moreover, our data suggest that regions within the C-terminus of Trio negatively regulate its interactions with Piccolo/Bassoon. These findings provide a mechanism for the presynaptic targeting of Trio and support a model in which Piccolo and Bassoon play a role in regulating neurotransmission through interactions with proteins, including Trio, that modulate the dynamic assembly of F-actin during cycles of synaptic vesicle exo- and endocytosis.
Collapse
Affiliation(s)
- Ryan T. Terry-Lorenzo
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Viviana I. Torres
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile, Alameda, Santiago, Chile
| | - Dhananjay Wagh
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Jose Galaz
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Selene K. Swanson
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Laurence Florens
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Michael P. Washburn
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Clarissa L. Waites
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Eckart D. Gundelfinger
- Dept. of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Richard J. Reimer
- Dept. of Neurology and Neurological Sciences Stanford University and Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Craig C. Garner
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
- German Centers for Neurodegenerative Diseases, Charité - Medical University, Berlin, Germany
- * E-mail:
| |
Collapse
|
47
|
Pacheco A, Gallo G. Actin filament-microtubule interactions in axon initiation and branching. Brain Res Bull 2016; 126:300-310. [PMID: 27491623 DOI: 10.1016/j.brainresbull.2016.07.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/28/2016] [Accepted: 07/30/2016] [Indexed: 10/21/2022]
Abstract
Neurons begin life as spherical cells. A major hallmark of neuronal development is the formation of elongating processes from the cell body which subsequently differentiate into dendrites and the axon. The formation and later development of neuronal processes is achieved through the concerted organization of actin filaments and microtubules. Here, we review the literature regarding recent advances in the understanding of cytoskeletal interactions in neurons focusing on the initiation of processes from neuronal cell bodies and the collateral branching of axons. The complex crosstalk between cytoskeletal elements is mediated by a cohort of proteins that either bind both cytoskeletal systems or allow one to regulate the other. Recent studies have highlighted the importance of microtubule plus-tip proteins in the regulation of the dynamics and organization of actin filaments, while also providing a mechanism for the subcellular capture and guidance of microtubule tips by actin filaments. Although the understanding of cytoskeletal crosstalk and interactions in neuronal morphogenesis has advanced significantly in recent years the appreciation of the neuron as an integrated cytoskeletal system remains a frontier.
Collapse
Affiliation(s)
- Almudena Pacheco
- Temple University, Lewis Kats School of Medicine, Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, 3500 North Broad Street, Philadelphia, PA 19140, United States
| | - Gianluca Gallo
- Temple University, Lewis Kats School of Medicine, Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, 3500 North Broad Street, Philadelphia, PA 19140, United States.
| |
Collapse
|
48
|
Pengelly RJ, Greville-Heygate S, Schmidt S, Seaby EG, Jabalameli MR, Mehta SG, Parker MJ, Goudie D, Fagotto-Kaufmann C, Mercer C, Debant A, Ennis S, Baralle D. Mutations specific to the Rac-GEF domain of TRIO cause intellectual disability and microcephaly. J Med Genet 2016; 53:735-742. [PMID: 27418539 PMCID: PMC5264232 DOI: 10.1136/jmedgenet-2016-103942] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 11/03/2022]
Abstract
BACKGROUND Neurodevelopmental disorders have challenged clinical genetics for decades, with over 700 genes implicated and many whose function remains unknown. The application of whole-exome sequencing is proving pivotal in closing the genotype/phenotype gap through the discovery of new genes and variants that help to unravel the pathogenic mechanisms driving neuropathogenesis. One such discovery includes TRIO, a gene recently implicated in neurodevelopmental delay. Trio is a Dbl family guanine nucleotide exchange factor (GEF) and a major regulator of neuronal development, controlling actin cytoskeleton dynamics by activating the GTPase Rac1. METHODS Whole-exome sequencing was undertaken on a family presenting with global developmental delay, microcephaly and mild dysmorphism. Father/daughter exome analysis was performed, followed by confirmatory Sanger sequencing and segregation analysis on four individuals. Three further patients were recruited through the deciphering developmental disorders (DDD) study. Functional studies were undertaken using patient-specific Trio protein mutations. RESULTS We identified a frameshift deletion in TRIO that segregated autosomal dominantly. By scrutinising data from DDD, we further identified three unrelated children with a similar phenotype who harboured de novo missense mutations in TRIO. Biochemical studies demonstrated that in three out of four families, the Trio mutations led to a markedly reduced Rac1 activation. CONCLUSIONS We describe an inherited global developmental delay phenotype associated with a frameshift deletion in TRIO. Additionally, we identify pathogenic de novo missense mutations in TRIO associated with the same consistent phenotype, intellectual disability, microcephaly and dysmorphism with striking digital features. We further functionally validate the importance of the GEF domain in Trio protein function. Our study demonstrates how genomic technologies are yet again proving prolific in diagnosing and advancing the understanding of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Reuben J Pengelly
- Department of Human Genetics and Genomic Medicine, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Susanne Schmidt
- Centre de Recherche en Biologie Cellulaire de Montpellier, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, Cédex, France
| | - Eleanor G Seaby
- Department of Human Genetics and Genomic Medicine, Faculty of Medicine, University of Southampton, Southampton, UK
| | - M Reza Jabalameli
- Department of Human Genetics and Genomic Medicine, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sarju G Mehta
- Department of Clinical Genetics, Cambridge University Hospital Trust, Cambridge, UK
| | - Michael J Parker
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital NHS Foundation Trust, OPD2, Northern General Hospital, Sheffield, UK
| | - David Goudie
- Department of Clinical Genetics, Ninewells Hospital, Dundee, UK
| | - Christine Fagotto-Kaufmann
- Centre de Recherche en Biologie Cellulaire de Montpellier, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, Cédex, France
| | - Catherine Mercer
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | | | - Anne Debant
- Centre de Recherche en Biologie Cellulaire de Montpellier, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, Cédex, France
| | - Sarah Ennis
- Department of Human Genetics and Genomic Medicine, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Diana Baralle
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| |
Collapse
|
49
|
GEFs and Rac GTPases control directional specificity of neurite extension along the anterior-posterior axis. Proc Natl Acad Sci U S A 2016; 113:6973-8. [PMID: 27274054 DOI: 10.1073/pnas.1607179113] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Although previous studies have identified many extracellular guidance molecules and intracellular signaling proteins that regulate axonal outgrowth and extension, most were conducted in the context of unidirectional neurite growth, in which the guidance cues either attract or repel growth cones. Very few studies addressed how intracellular signaling molecules differentially specify bidirectional outgrowth. Here, using the bipolar PLM neurons in Caenorhabditis elegans, we show that the guanine nucleotide exchange factors (GEFs) UNC-73/Trio and TIAM-1 promote anterior and posterior neurite extension, respectively. The Rac subfamily GTPases act downstream of the GEFs; CED-10/Rac1 is activated by TIAM-1, whereas CED-10 and MIG-2/RhoG act redundantly downstream of UNC-73. Moreover, these two pathways antagonize each other and thus regulate the directional bias of neuritogenesis. Our study suggests that directional specificity of neurite extension is conferred through the intracellular activation of distinct GEFs and Rac GTPases.
Collapse
|
50
|
van de Willige D, Hoogenraad CC, Akhmanova A. Microtubule plus-end tracking proteins in neuronal development. Cell Mol Life Sci 2016; 73:2053-77. [PMID: 26969328 PMCID: PMC4834103 DOI: 10.1007/s00018-016-2168-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/04/2016] [Accepted: 02/22/2016] [Indexed: 11/28/2022]
Abstract
Regulation of the microtubule cytoskeleton is of pivotal importance for neuronal development and function. One such regulatory mechanism centers on microtubule plus-end tracking proteins (+TIPs): structurally and functionally diverse regulatory factors, which can form complex macromolecular assemblies at the growing microtubule plus-ends. +TIPs modulate important properties of microtubules including their dynamics and their ability to control cell polarity, membrane transport and signaling. Several neurodevelopmental and neurodegenerative diseases are associated with mutations in +TIPs or with misregulation of these proteins. In this review, we focus on the role and regulation of +TIPs in neuronal development and associated disorders.
Collapse
Affiliation(s)
- Dieudonnée van de Willige
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|