1
|
Faraji N, Ebadpour N, Abavisani M, Gorji A. Unlocking Hope: Therapeutic Advances and Approaches in Modulating the Wnt Pathway for Neurodegenerative Diseases. Mol Neurobiol 2025; 62:3630-3652. [PMID: 39313658 PMCID: PMC11790780 DOI: 10.1007/s12035-024-04462-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024]
Abstract
Neurodegenerative diseases (NDs) are conditions characterized by sensory, motor, and cognitive impairments due to alterations in the structure and function of neurons in the central nervous system (CNS). Despite their widespread occurrence, the exact causes of NDs remain largely elusive, and existing treatments fall short in efficacy. The Wnt signaling pathway is an emerging molecular pathway that has been linked to the development and progression of various NDs. Wnt signaling governs numerous cellular processes, such as survival, polarity, proliferation, differentiation, migration, and fate specification, via a complex network of proteins. In the adult CNS, Wnt signaling regulates synaptic transmission, plasticity, memory formation, neurogenesis, neuroprotection, and neuroinflammation, all essential for maintaining neuronal function and integrity. Dysregulation of both canonical and non-canonical Wnt signaling pathways contributes to neurodegeneration through various mechanisms, such as amyloid-β accumulation, tau protein hyperphosphorylation, dopaminergic neuron degeneration, and synaptic dysfunction, prompting investigations into Wnt modulation as a therapeutic target to restore neuronal function and prevent or delay neurodegenerative processes. Modulating Wnt signaling has the potential to restore neuronal function and impede or postpone neurodegenerative processes, offering a therapeutic approach for targeting NDs. In this article, the current knowledge about how Wnt signaling works in Alzheimer's disease and Parkinson's disease is discussed. Our study aims to explore the molecular mechanisms, recent discoveries, and challenges involved in developing Wnt-based therapies.
Collapse
Affiliation(s)
- Navid Faraji
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Epilepsy Research Center, Münster University, Münster, Germany.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Neurosurgery Department, Münster University, Münster, Germany.
| |
Collapse
|
2
|
Wang Y, Liu X, Liu Z, Hua S, Jiang K. APC orchestrates microtubule dynamics by acting as a positive regulator of KIF2A and a negative regulator of CLASPs. CELL INSIGHT 2025; 4:100210. [PMID: 39640087 PMCID: PMC11617872 DOI: 10.1016/j.cellin.2024.100210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 12/07/2024]
Abstract
Tumor suppressor protein Adenomatous polyposis coli protein (APC) is an EB-binding and microtubule (MT) plus end-tracking protein; however, how exactly APC regulates MT dynamics remains elusive. Here, we show that in LLC-PK1 cells, APC and KIF2A, an MT depolymerase, form a complex clustering at the cell edge and destabilize MTs at the MT plus ends. Further biochemical characterization and mutational analysis reveal key residues for the APC-KIF2A interaction. In addition, APC counteracts the major MT-stabilizer CLASPs at MT plus ends and promotes directional cell migration via modulating cell adhesion force. Reconstitution experiments demonstrate that APC potentiates KIF2A-induced MT catastrophes and antagonizes the stabilizing effect of CLASP2 in vitro. In summary, APC functions as a positive regulator of MT-destabilizer and a negative regulator of MT-stabilizer to orchestrate MT dynamics.
Collapse
Affiliation(s)
- Yong Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Xinping Liu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Zheng Liu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Shasha Hua
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Kai Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
3
|
Chen YJ, Tseng SC, Chen PT, Hwang E. The non-mitotic role of HMMR in regulating the localization of TPX2 and the dynamics of microtubules in neurons. eLife 2024; 13:RP94547. [PMID: 38904660 PMCID: PMC11192530 DOI: 10.7554/elife.94547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
A functional nervous system is built upon the proper morphogenesis of neurons to establish the intricate connection between them. The microtubule cytoskeleton is known to play various essential roles in this morphogenetic process. While many microtubule-associated proteins (MAPs) have been demonstrated to participate in neuronal morphogenesis, the function of many more remains to be determined. This study focuses on a MAP called HMMR in mice, which was originally identified as a hyaluronan binding protein and later found to possess microtubule and centrosome binding capacity. HMMR exhibits high abundance on neuronal microtubules and altering the level of HMMR significantly affects the morphology of neurons. Instead of confining to the centrosome(s) like cells in mitosis, HMMR localizes to microtubules along axons and dendrites. Furthermore, transiently expressing HMMR enhances the stability of neuronal microtubules and increases the formation frequency of growing microtubules along the neurites. HMMR regulates the microtubule localization of a non-centrosomal microtubule nucleator TPX2 along the neurite, offering an explanation for how HMMR contributes to the promotion of growing microtubules. This study sheds light on how cells utilize proteins involved in mitosis for non-mitotic functions.
Collapse
Affiliation(s)
- Yi-Ju Chen
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Shun-Cheng Tseng
- Department of Orthopedic Surgery, Changhua Christian HospitalChanghuaTaiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Peng-Tzu Chen
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Eric Hwang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| |
Collapse
|
4
|
Huang H, Majumder T, Khot B, Suriyaarachchi H, Yang T, Shao Q, Tirukovalluru S, Liu G. The role of microtubule-associated protein tau in netrin-1 attractive signaling. J Cell Sci 2024; 137:jcs261244. [PMID: 38197773 PMCID: PMC10906489 DOI: 10.1242/jcs.261244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/24/2023] [Indexed: 01/11/2024] Open
Abstract
Direct binding of netrin receptors with dynamic microtubules (MTs) in the neuronal growth cone plays an important role in netrin-mediated axon guidance. However, how netrin-1 (NTN1) regulates MT dynamics in axon turning remains a major unanswered question. Here, we show that the coupling of netrin-1 receptor DCC with tau (MAPT)-regulated MTs is involved in netrin-1-promoted axon attraction. Tau directly interacts with DCC and partially overlaps with DCC in the growth cone of primary neurons. Netrin-1 induces this interaction and the colocalization of DCC and tau in the growth cone. The netrin-1-induced interaction of tau with DCC relies on MT dynamics and TUBB3, a highly dynamic β-tubulin isotype in developing neurons. Netrin-1 increased cosedimentation of DCC with tau and TUBB3 in MTs, and knockdown of either tau or TUBB3 mutually blocked this effect. Downregulation of endogenous tau levels by tau shRNAs inhibited netrin-1-induced axon outgrowth, branching and commissural axon attraction in vitro, and led to defects in spinal commissural axon projection in vivo. These findings suggest that tau is a key MT-associated protein coupling DCC with MT dynamics in netrin-1-promoted axon attraction.
Collapse
Affiliation(s)
- Huai Huang
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Bhakti Khot
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Harindi Suriyaarachchi
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Shraddha Tirukovalluru
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| |
Collapse
|
5
|
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
|
6
|
Anand AA, Khan M, V M, Kar D. The Molecular Basis of Wnt/ β-Catenin Signaling Pathways in Neurodegenerative Diseases. Int J Cell Biol 2023; 2023:9296092. [PMID: 37780577 PMCID: PMC10539095 DOI: 10.1155/2023/9296092] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 10/03/2023] Open
Abstract
Defective Wnt signaling is found to be associated with various neurodegenerative diseases. In the canonical pathway, the Frizzled receptor (Fzd) and the lipoprotein receptor-related proteins 5/6 (LRP5/LRP6) create a seven-pass transmembrane receptor complex to which the Wnt ligands bind. This interaction causes the tumor suppressor adenomatous polyposis coli gene product (APC), casein kinase 1 (CK1), and GSK-3β (glycogen synthase kinase-3 beta) to be recruited by the scaffold protein Dishevelled (Dvl), which in turn deactivates the β-catenin destruction complex. This inactivation stops the destruction complex from phosphorylating β-catenin. As a result, β-catenin first builds up in the cytoplasm and then migrates into the nucleus, where it binds to the Lef/Tcf transcription factor to activate the transcription of more than 50 Wnt target genes, including those involved in cell growth, survival, differentiation, neurogenesis, and inflammation. The treatments that are currently available for neurodegenerative illnesses are most commonly not curative in nature but are only symptomatic. According to all available research, restoring Wnt/β-catenin signaling in the brains of patients with neurodegenerative disorders, particularly Alzheimer's and Parkinson's disease, would improve the condition of several patients with neurological disorders. The importance of Wnt activators and modulators in patients with such illnesses is to mainly restore rather than overstimulate the Wnt/β-catenin signaling, thereby reestablishing the equilibrium between Wnt-OFF and Wnt-ON states. In this review, we have tried to summarize the significance of the Wnt canonical pathway in the pathophysiology of certain neurodegenerative diseases, such as Alzheimer's disease, cerebral ischemia, Parkinson's disease, Huntington's disease, multiple sclerosis, and other similar diseases, and as to how can it be restored in these patients.
Collapse
Affiliation(s)
- Ananya Anurag Anand
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad 211012, India
| | - Misbah Khan
- Department of Biotechnology, Ramaiah University of Applied Sciences, Bengaluru 560054, India
| | - Monica V
- Department of Biotechnology, Ramaiah University of Applied Sciences, Bengaluru 560054, India
| | - Debasish Kar
- Department of Biotechnology, Ramaiah University of Applied Sciences, Bengaluru 560054, India
| |
Collapse
|
7
|
Atkins M, Nicol X, Fassier C. Microtubule remodelling as a driving force of axon guidance and pruning. Semin Cell Dev Biol 2023; 140:35-53. [PMID: 35710759 DOI: 10.1016/j.semcdb.2022.05.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023]
Abstract
The establishment of neuronal connectivity relies on the microtubule (MT) cytoskeleton, which provides mechanical support, roads for axonal transport and mediates signalling events. Fine-tuned spatiotemporal regulation of MT functions by tubulin post-translational modifications and MT-associated proteins is critical for the coarse wiring and subsequent refinement of neuronal connectivity. The defective regulation of these processes causes a wide range of neurodevelopmental disorders associated with connectivity defects. This review focuses on recent studies unravelling how MT composition, post-translational modifications and associated proteins influence MT functions in axon guidance and/or pruning to build functional neuronal circuits. We here summarise experimental evidence supporting the key role of this network as a driving force for growth cone steering and branch-specific axon elimination. We further provide a global overview of the MT-interactors that tune developing axon behaviours, with a special emphasis on their emerging versatility in the regulation of MT dynamics/structure. Recent studies establishing the key and highly selective role of the tubulin code in the regulation of MT functions in axon pathfinding are also reported. Finally, our review highlights the emerging molecular links between these MT regulation processes and guidance signals that wire the nervous system.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM, UMR-S 1270, Institut du Fer à Moulin, Sorbonne Université, F-75005 Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Coralie Fassier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France.
| |
Collapse
|
8
|
Triantopoulou N, Vidaki M. Local mRNA translation and cytoskeletal reorganization: Mechanisms that tune neuronal responses. Front Mol Neurosci 2022; 15:949096. [PMID: 35979146 PMCID: PMC9376447 DOI: 10.3389/fnmol.2022.949096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/07/2022] [Indexed: 12/31/2022] Open
Abstract
Neurons are highly polarized cells with significantly long axonal and dendritic extensions that can reach distances up to hundreds of centimeters away from the cell bodies in higher vertebrates. Their successful formation, maintenance, and proper function highly depend on the coordination of intricate molecular networks that allow axons and dendrites to quickly process information, and respond to a continuous and diverse cascade of environmental stimuli, often without enough time for communication with the soma. Two seemingly unrelated processes, essential for these rapid responses, and thus neuronal homeostasis and plasticity, are local mRNA translation and cytoskeletal reorganization. The axonal cytoskeleton is characterized by high stability and great plasticity; two contradictory attributes that emerge from the powerful cytoskeletal rearrangement dynamics. Cytoskeletal reorganization is crucial during nervous system development and in adulthood, ensuring the establishment of proper neuronal shape and polarity, as well as regulating intracellular transport and synaptic functions. Local mRNA translation is another mechanism with a well-established role in the developing and adult nervous system. It is pivotal for axonal guidance and arborization, synaptic formation, and function and seems to be a key player in processes activated after neuronal damage. Perturbations in the regulatory pathways of local translation and cytoskeletal reorganization contribute to various pathologies with diverse clinical manifestations, ranging from intellectual disabilities (ID) to autism spectrum disorders (ASD) and schizophrenia (SCZ). Despite the fact that both processes are essential for the orchestration of pathways critical for proper axonal and dendritic function, the interplay between them remains elusive. Here we review our current knowledge on the molecular mechanisms and specific interaction networks that regulate and potentially coordinate these interconnected processes.
Collapse
Affiliation(s)
- Nikoletta Triantopoulou
- Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
| | - Marina Vidaki
- Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
- *Correspondence: Marina Vidaki,
| |
Collapse
|
9
|
Upadhyay J, Ansari MN, Samad A, Sayana A. Dysregulation of multiple signaling pathways: A possible cause of cerebral palsy. Exp Biol Med (Maywood) 2022; 247:779-787. [PMID: 35253451 DOI: 10.1177/15353702221081022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cerebral palsy (CP) is a lifelong disability characterized by the impairment of brain functions that result in improper posture and abnormal motor patterns. Understanding this brain abnormality and the role of genetic, epigenetic, and non-genetic factors such as signaling pathway dysregulation and cytokine dysregulation in the pathogenesis of CP is a complex process. Hypoxic-ischemic injury and prematurity are two well-known contributors of CP. Like in the case of other neurodevelopmental disorders such as intellectual disability and autism, the genomic constituents in CP are highly complex. The neuroinflammation that is triggered by maternal cytokine response plays a critical role in the pathogenesis of fetal inflammation response, which is one of the contributing factors of CP, and it continues even after the birth of children suffering from CP. Canonical Wnt signaling pathway is important for the development of mammalian fetal brain and it regulates distinct processes including neurogenesis. The glycogen synthase kinase-3 (GSK-3) antagonistic activity in the Wnt signaling pathway plays a crucial role in neurogenesis and neural development. In this review, we investigated several genetic and non-genetic pathways that are involved in the pathogenesis of CP and their regulation, impairment, and implications for causing CP during embryonic growth and developmental period. Investigating the role of these pathways help to develop novel therapeutic interventions and biomarkers for early diagnosis and treatment. This review also helps us to comprehend the mechanical approach of various signaling pathways, as well as their consequences and relevance in the understanding of CP.
Collapse
Affiliation(s)
- Jyoti Upadhyay
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Mohd Nazam Ansari
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Abdul Samad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tishk International University, Erbil 44001, Iraq
| | | |
Collapse
|
10
|
Kimmerlin Q, Strassel C, Eckly A, Lanza F. The tubulin code in platelet biogenesis. Semin Cell Dev Biol 2022; 137:63-73. [PMID: 35148939 DOI: 10.1016/j.semcdb.2022.01.010] [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: 05/18/2021] [Revised: 01/12/2022] [Accepted: 01/31/2022] [Indexed: 11/28/2022]
Abstract
Blood platelets are small non-nucleated cellular fragments that prevent and stop hemorrhages. They are produced in the bone marrow by megakaryocytes through megakaryopoiesis. This intricate process involves profound microtubule rearrangements culminating in the formation of a unique circular sub-membranous microtubule array, the marginal band, which supports the typical disc-shaped morphology of platelets. Mechanistically, these processes are thought to be controlled by a specific tubulin code. In this review, we summarize the current knowledge on the key isotypes, notably β1-, α4A- and α8-tubulin, and putative post-translational modifications, involved in platelet and marginal band formation. Additionally, we provide a provisional list of microtubule-associated proteins (MAPs) involved in these processes and a survey of tubulin variants identified in patients presenting defective platelet production. A comprehensive characterization of the platelet tubulin code and the identification of essential MAPs may be expected in the near future to shed new light on a very specialized microtubule assembly process with applications in platelet diseases and transfusion.
Collapse
Affiliation(s)
- Quentin Kimmerlin
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Strasbourg, France.
| | - Catherine Strassel
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Strasbourg, France.
| | - Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Strasbourg, France.
| | - François Lanza
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Strasbourg, France.
| |
Collapse
|
11
|
Sánchez-Huertas C, Herrera E. With the Permission of Microtubules: An Updated Overview on Microtubule Function During Axon Pathfinding. Front Mol Neurosci 2021; 14:759404. [PMID: 34924953 PMCID: PMC8675249 DOI: 10.3389/fnmol.2021.759404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/27/2023] Open
Abstract
During the establishment of neural circuitry axons often need to cover long distances to reach remote targets. The stereotyped navigation of these axons defines the connectivity between brain regions and cellular subtypes. This chemotrophic guidance process mostly relies on the spatio-temporal expression patterns of extracellular proteins and the selective expression of their receptors in projection neurons. Axon guidance is stimulated by guidance proteins and implemented by neuronal traction forces at the growth cones, which engage local cytoskeleton regulators and cell adhesion proteins. Different layers of guidance signaling regulation, such as the cleavage and processing of receptors, the expression of co-receptors and a wide variety of intracellular cascades downstream of receptors activation, have been progressively unveiled. Also, in the last decades, the regulation of microtubule (MT) assembly, stability and interactions with the submembranous actin network in the growth cone have emerged as crucial effector mechanisms in axon pathfinding. In this review, we will delve into the intracellular signaling cascades downstream of guidance receptors that converge on the MT cytoskeleton of the growing axon. In particular, we will focus on the microtubule-associated proteins (MAPs) network responsible of MT dynamics in the axon and growth cone. Complementarily, we will discuss new evidences that connect defects in MT scaffold proteins, MAPs or MT-based motors and axon misrouting during brain development.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Alicante, Spain
| | | |
Collapse
|
12
|
Prasad R, Jung H, Tan A, Song Y, Moon S, Shaker MR, Sun W, Lee J, Ryu H, Lim HK, Jho EH. Hypermethylation of Mest promoter causes aberrant Wnt signaling in patients with Alzheimer's disease. Sci Rep 2021; 11:20075. [PMID: 34625606 PMCID: PMC8501037 DOI: 10.1038/s41598-021-99562-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/28/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and behavioral changes. Extracellular deposition of amyloid plaques (Aβ) and intracellular deposition of neurofibrillary tangles in neurons are the major pathogenicities of AD. However, drugs targeting these therapeutic targets are not effective. Therefore, novel targets for the treatment of AD urgently need to be identified. Expression of the mesoderm-specific transcript (Mest) is regulated by genomic imprinting, where only the paternal allele is active for transcription. We identified hypermethylation on the Mest promoter, which led to a reduction in Mest mRNA levels and activation of Wnt signaling in brain tissues of AD patients. Mest knockout (KO) using the CRIPSR/Cas9 system in mouse embryonic stem cells and P19 embryonic carcinoma cells leads to neuronal differentiation arrest. Depletion of Mest in primary hippocampal neurons via lentivirus expressing shMest or inducible KO system causes neurodegeneration. Notably, depletion of Mest in primary cortical neurons of rats leads to tau phosphorylation at the S199 and T231 sites. Overall, our data suggest that hypermethylation of the Mest promoter may cause or facilitate the progression of AD.
Collapse
Affiliation(s)
- Renuka Prasad
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Hwajin Jung
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Anderson Tan
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Yonghee Song
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Sungho Moon
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Mohammed R Shaker
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Junghee Lee
- Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hoon Ryu
- Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Hyun Kook Lim
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea.
| |
Collapse
|
13
|
Puri D, Ponniah K, Biswas K, Basu A, Dey S, Lundquist EA, Ghosh-Roy A. Wnt signaling establishes the microtubule polarity in neurons through regulation of Kinesin-13. J Cell Biol 2021; 220:212396. [PMID: 34137792 DOI: 10.1083/jcb.202005080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Neuronal polarization is facilitated by the formation of axons with parallel arrays of plus-end-out and dendrites with the nonuniform orientation of microtubules. In C. elegans, the posterior lateral microtubule (PLM) neuron is bipolar with its two processes growing along the anterior-posterior axis under the guidance of Wnt signaling. Here we found that loss of the Kinesin-13 family microtubule-depolymerizing enzyme KLP-7 led to the ectopic extension of axon-like processes from the PLM cell body. Live imaging of the microtubules and axonal transport revealed mixed polarity of the microtubules in the short posterior process, which is dependent on both KLP-7 and the minus-end binding protein PTRN-1. KLP-7 is positively regulated in the posterior process by planar cell polarity components of Wnt involving rho-1/rock to induce mixed polarity of microtubules, whereas it is negatively regulated in the anterior process by the unc-73/ced-10 cascade to establish a uniform microtubule polarity. Our work elucidates how evolutionarily conserved Wnt signaling establishes the microtubule polarity in neurons through Kinesin-13.
Collapse
Affiliation(s)
- Dharmendra Puri
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Keerthana Ponniah
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Kasturi Biswas
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Atrayee Basu
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Swagata Dey
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Erik A Lundquist
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS
| | - Anindya Ghosh-Roy
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| |
Collapse
|
14
|
Pinto MJ, Tomé D, Almeida RD. The Ubiquitinated Axon: Local Control of Axon Development and Function by Ubiquitin. J Neurosci 2021; 41:2796-2813. [PMID: 33789876 PMCID: PMC8018891 DOI: 10.1523/jneurosci.2251-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 02/01/2023] Open
Abstract
Ubiquitin tagging sets protein fate. With a wide range of possible patterns and reversibility, ubiquitination can assume many shapes to meet specific demands of a particular cell across time and space. In neurons, unique cells with functionally distinct axons and dendrites harboring dynamic synapses, the ubiquitin code is exploited at the height of its power. Indeed, wide expression of ubiquitination and proteasome machinery at synapses, a diverse brain ubiquitome, and the existence of ubiquitin-related neurodevelopmental diseases support a fundamental role of ubiquitin signaling in the developing and mature brain. While special attention has been given to dendritic ubiquitin-dependent control, how axonal biology is governed by this small but versatile molecule has been considerably less discussed. Herein, we set out to explore the ubiquitin-mediated spatiotemporal control of an axon's lifetime: from its differentiation and growth through presynaptic formation, function, and pruning.
Collapse
Affiliation(s)
- Maria J Pinto
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Diogo Tomé
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ramiro D Almeida
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, 3810-193, Portugal
| |
Collapse
|
15
|
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: 23] [Impact Index Per Article: 5.8] [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
|
16
|
Morenilla-Palao C, López-Cascales MT, López-Atalaya JP, Baeza D, Calvo-Díaz L, Barco A, Herrera E. A Zic2-regulated switch in a noncanonical Wnt/βcatenin pathway is essential for the formation of bilateral circuits. SCIENCE ADVANCES 2020; 6:6/46/eaaz8797. [PMID: 33188033 PMCID: PMC7673756 DOI: 10.1126/sciadv.aaz8797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 09/30/2020] [Indexed: 05/06/2023]
Abstract
The Wnt pathway is involved in a wide array of biological processes during development and is deregulated in many pathological scenarios. In neurons, Wnt proteins promote both axon extension and repulsion, but the molecular mechanisms underlying these opposing axonal responses are unknown. Here, we show that Wnt5a is expressed at the optic chiasm midline and promotes the crossing of retinal axons by triggering an alternative Wnt pathway that depends on the accumulation of βcatenin but does not activate the canonical pathway. In ipsilateral neurons, the transcription factor Zic2 switches this alternative Wnt pathway by regulating the expression of a set of Wnt receptors and intracellular proteins. In combination with this alternative Wnt pathway, the asymmetric activation of EphB1 receptors at the midline phosphorylates βcatenin and elicits a repulsive response. This alternative Wnt pathway and its Zic2-triggered switch may operate in other contexts that require a two-way response to Wnt ligands.
Collapse
Affiliation(s)
- Cruz Morenilla-Palao
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - María Teresa López-Cascales
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - José P López-Atalaya
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Diana Baeza
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Luís Calvo-Díaz
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Angel Barco
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Eloísa Herrera
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain.
| |
Collapse
|
17
|
Silencing Celsr2 inhibits the proliferation and migration of Schwann cells through suppressing the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2020; 533:623-630. [PMID: 32988580 DOI: 10.1016/j.bbrc.2020.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 01/26/2023]
Abstract
After a peripheral nerve injury, the remaining Schwann cells undergo proliferation and adopt a migratory phenotype to prepare for the regeneration of nerves. Celsr2 has been reported to play an important role in the development and maintenance of the function of the nervous system. However, the role and mechanism of Celsr2 during peripheral nerve regeneration remain unknown. Here, we showed that after sciatic nerve injury, Celsr2 mRNA and protein were significantly increased in nerve tissues. In addition, silencing Celsr2 decreased the ki67-positve portion and the migration distance of Schwann cells in vivo. In vitro, the results of MTT and EdU staining, transwell and wound healing assays indicated that Celsr2 siRNA-transfected primary Schwann cells showed significant decrease in proliferation and migration compared to that seen in negative control (NC)-transfected cells. Furthermore, we found that Wnt/β-catenin luciferase activity was reduced, as were the expression of β-catenin in the nucleus and the mRNA levels of its downstream genes Cyclin D1 and MMP-7 in Celsr2 siRNA-transfected primary Schwann cells. Further investigations showed that silencing Celsr2 inhibited the phosphorylation of GSK3β. Moreover, specific activators of the Wnt/β-catenin pathway, LiCl or mutant β-catenin (S33Y), partially reversed the inhibitory effect of Celsr2 siRNA. Taken together, our data indicated that silencing Celsr2 inhibited Schwann cells migration and proliferation through the suppressing Wnt/β-catenin pathway, providing a potential target for peripheral nerve regeneration.
Collapse
|
18
|
Coullery R, Pacchioni AM, Rosso SB. Exposure to glyphosate during pregnancy induces neurobehavioral alterations and downregulation of Wnt5a-CaMKII pathway. Reprod Toxicol 2020; 96:390-398. [PMID: 32805371 DOI: 10.1016/j.reprotox.2020.08.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 01/08/2023]
Abstract
Glyphosate-based formulations are the most popular herbicide used around the world. These herbicides are widely applied in agriculture to control weeds on genetically modified crops. Although there is much evidence showing that glyphosate-based herbicides induce toxic effect on reproductive and hepatic systems, and also cause oxidative damage on cells, studies from recent years revealed that the nervous system may represent a key target for their toxicity. In the present work, we evaluated the effect of glyphosate (without adjuvants) in neonate rats after gestational exposure. Particularly, we examined whether glyphosate during gestation affected the nervous system function at early development. Pregnant Wistar rats were treated with 24 or 35 mg/kg of pure glyphosate every 48 h and neurobehavioral studies were performed. Our results indicated that gestational exposure to glyphosate induced changes in reflexes development, motor activity and cognitive function, in a dose-dependent manner. To go further, we evaluated whether prenatal exposure to glyphosate affected the Ca+2-mediated Wnt non-canonical signaling pathway. Results indicated that embryos exposed to glyphosate showed an inhibition of Wnt5a-CaMKII signaling pathway, an essential cascade controlling the formation and integration of neural circuits. Taken together, these findings suggest that gestational exposure to glyphosate leads to a downregulation of Wnt/Ca+2 pathway that could induce a developmental neurotoxicity evidenced by deficits at behavioral and cognitive levels in rat pups.
Collapse
Affiliation(s)
- Romina Coullery
- Área Toxicología, Departamento de Ciencias de los Alimentos y Medio Ambiente, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha 531, S2002LRK Rosario, Santa Fe, Argentina
| | - Alejandra M Pacchioni
- Área Toxicología, Departamento de Ciencias de los Alimentos y Medio Ambiente, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha 531, S2002LRK Rosario, Santa Fe, Argentina
| | - Silvana B Rosso
- Área Toxicología, Departamento de Ciencias de los Alimentos y Medio Ambiente, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha 531, S2002LRK Rosario, Santa Fe, Argentina.
| |
Collapse
|
19
|
Turner-Bridger B, Caterino C, Cioni JM. Molecular mechanisms behind mRNA localization in axons. Open Biol 2020; 10:200177. [PMID: 32961072 PMCID: PMC7536069 DOI: 10.1098/rsob.200177] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
Messenger RNA (mRNA) localization allows spatiotemporal regulation of the proteome at the subcellular level. This is observed in the axons of neurons, where mRNA localization is involved in regulating neuronal development and function by orchestrating rapid adaptive responses to extracellular cues and the maintenance of axonal homeostasis through local translation. Here, we provide an overview of the key findings that have broadened our knowledge regarding how specific mRNAs are trafficked and localize to axons. In particular, we review transcriptomic studies investigating mRNA content in axons and the molecular principles underpinning how these mRNAs arrived there, including cis-acting mRNA sequences and trans-acting proteins playing a role. Further, we discuss evidence that links defective axonal mRNA localization and pathological outcomes.
Collapse
Affiliation(s)
- Benita Turner-Bridger
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, UK
| | - Cinzia Caterino
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Jean-Michel Cioni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| |
Collapse
|
20
|
Drechsler M, Lang LF, Al-Khatib L, Dirks H, Burger M, Schönlieb CB, Palacios IM. Optical flow analysis reveals that Kinesin-mediated advection impacts the orientation of microtubules in the Drosophila oocyte. Mol Biol Cell 2020; 31:1246-1258. [PMID: 32267197 PMCID: PMC7353148 DOI: 10.1091/mbc.e19-08-0440] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The orientation of microtubule (MT) networks is exploited by motors to deliver cargoes to specific intracellular destinations and is thus essential for cell polarity and function. Reconstituted in vitro systems have largely contributed to understanding the molecular framework regulating the behavior of MT filaments. In cells, however, MTs are exposed to various biomechanical forces that might impact on their orientation, but little is known about it. Oocytes, which display forceful cytoplasmic streaming, are excellent model systems to study the impact of motion forces on cytoskeletons in vivo. Here we implement variational optical flow analysis as a new approach to analyze the polarity of MTs in the Drosophila oocyte, a cell that displays distinct Kinesin-dependent streaming. After validating the method as robust for describing MT orientation from confocal movies, we find that increasing the speed of flows results in aberrant plus end growth direction. Furthermore, we find that in oocytes where Kinesin is unable to induce cytoplasmic streaming, the growth direction of MT plus ends is also altered. These findings lead us to propose that cytoplasmic streaming - and thus motion by advection – contributes to the correct orientation of MTs in vivo. Finally, we propose a possible mechanism for a specialized cytoplasmic actin network (the actin mesh) to act as a regulator of flow speeds to counteract the recruitment of Kinesin to MTs.
Collapse
Affiliation(s)
- Maik Drechsler
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK.,Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, United Kingdom.,Department of Zoology and Developmental Biology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Lukas F Lang
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge CB3 0WA, United Kingdom
| | - Layla Al-Khatib
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Hendrik Dirks
- Institute for Computational and Applied Mathematics, University of Münster, 48149 Münster, Germany
| | - Martin Burger
- Department of Mathematics, Friedrich-Alexander Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Carola-Bibiane Schönlieb
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge CB3 0WA, United Kingdom
| | - Isabel M Palacios
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK.,Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, United Kingdom
| |
Collapse
|
21
|
Weiner AT, Seebold DY, Torres-Gutierrez P, Folker C, Swope RD, Kothe GO, Stoltz JG, Zalenski MK, Kozlowski C, Barbera DJ, Patel MA, Thyagarajan P, Shorey M, Nye DMR, Keegan M, Behari K, Song S, Axelrod JD, Rolls MM. Endosomal Wnt signaling proteins control microtubule nucleation in dendrites. PLoS Biol 2020; 18:e3000647. [PMID: 32163403 PMCID: PMC7067398 DOI: 10.1371/journal.pbio.3000647] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/07/2020] [Indexed: 12/21/2022] Open
Abstract
Dendrite microtubules are polarized with minus-end-out orientation in Drosophila neurons. Nucleation sites concentrate at dendrite branch points, but how they localize is not known. Using Drosophila, we found that canonical Wnt signaling proteins regulate localization of the core nucleation protein γTubulin (γTub). Reduction of frizzleds (fz), arrow (low-density lipoprotein receptor-related protein [LRP] 5/6), dishevelled (dsh), casein kinase Iγ, G proteins, and Axin reduced γTub-green fluorescent protein (GFP) at branch points, and two functional readouts of dendritic nucleation confirmed a role for Wnt signaling proteins. Both dsh and Axin localized to branch points, with dsh upstream of Axin. Moreover, tethering Axin to mitochondria was sufficient to recruit ectopic γTub-GFP and increase microtubule dynamics in dendrites. At dendrite branch points, Axin and dsh colocalized with early endosomal marker Rab5, and new microtubule growth initiated at puncta marked with fz, dsh, Axin, and Rab5. We propose that in dendrites, canonical Wnt signaling proteins are housed on early endosomes and recruit nucleation sites to branch points.
Collapse
Affiliation(s)
- Alexis T. Weiner
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Dylan Y. Seebold
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Pedro Torres-Gutierrez
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Christin Folker
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Rachel D. Swope
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Gregory O. Kothe
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Jessica G. Stoltz
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Madeleine K. Zalenski
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Christopher Kozlowski
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Dylan J. Barbera
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Mit A. Patel
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Pankajam Thyagarajan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Matthew Shorey
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Derek M. R. Nye
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Matthew Keegan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Kana Behari
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Song Song
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Jeffrey D. Axelrod
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Melissa M. Rolls
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
22
|
Shao Q, Yang T, Huang H, Majumder T, Khot BA, Khouzani MM, Alarmanazi F, Gore YK, Liu G. Disease-associated mutations in human TUBB3 disturb netrin repulsive signaling. PLoS One 2019; 14:e0218811. [PMID: 31226147 PMCID: PMC6588280 DOI: 10.1371/journal.pone.0218811] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/10/2019] [Indexed: 11/28/2022] Open
Abstract
Missense mutations in the human TUBB3 gene cause a variety of neurological disorders associated with defects in axon guidance and neuronal migration, but the underlying molecular mechanisms are not well understood. Recent studies have shown that direct coupling of dynamic TUBB3 in microtubules with netrin receptors is required for netrin-1-mediated axon guidance, and the interaction of netrin-1 repulsive receptor UNC5C with TUBB3 is involved in netrin-1 mediated axonal repulsion. Here, we report that TUBB3 mutations perturb netrin-1/UNC5C repulsive signaling in the developing nervous system. Among twelve mutants reported in previous studies, five of them show significantly reduced interaction with UNC5C in comparison to the wild-type TUBB3. TUBB3 mutants R262C and R62Q exhibit decreased subcellular colocalization with UNC5C in the peripheral area of the growth cone of primary mouse neurons. Netrin-1 reduces the colocalization of UNC5C with wild-type TUBB3, but not TUBB3 mutants R262C or R62Q, in the growth cone. Results from the in vitro cosedimentation assay indicate that netrin-1 inhibits cosedimentation of UNC5C with polymerized microtubules in primary mouse neurons expressing the wild-type TUBB3, but not R262C or R62Q. Expression of either R262C or R62Q not only blocks netrin-1-induced growth cone collapse and axonal repulsion of primary EGL cells in vitro, but also results in axon projections defects of chicken dorsal root ganglion neurons in ovo. Our study reveals that human TUBB3 mutations specifically perturb netrin-1/UNC5C-mediated repulsion.
Collapse
Affiliation(s)
- Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Huai Huang
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Bhakti Ajit Khot
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | | | - Farrah Alarmanazi
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Yasmin K. Gore
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
- * E-mail:
| |
Collapse
|
23
|
Stanganello E, Zahavi EE, Burute M, Smits J, Jordens I, Maurice MM, Kapitein LC, Hoogenraad CC. Wnt Signaling Directs Neuronal Polarity and Axonal Growth. iScience 2019; 13:318-327. [PMID: 30878878 PMCID: PMC6423405 DOI: 10.1016/j.isci.2019.02.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 09/18/2018] [Accepted: 02/26/2019] [Indexed: 11/19/2022] Open
Abstract
The establishment of neuronal polarity is driven by cytoskeletal remodeling that stabilizes and promotes the growth of a single axon from one of the multiple neurites. The importance of the local microtubule stabilization in this process has been revealed however, the external signals initiating the cytoskeletal rearrangements are not completely understood. In this study, we show that local activation of the canonical Wnt pathway regulates neuronal polarity and axonal outgrowth. We found that in the early stages of neuronal polarization, Wnt3a accumulates in one of the neurites of unpolarized cells and thereby could determine axon positioning. Subsequently, Wnt3a localizes to the growing axon, where it activates the canonical Wnt pathway and controls axon positioning and axonal length. We propose a model in which Wnt3a regulates the formation and growth of the axon by activating local intracellular signaling events leading to microtubule remodeling. Wnt3a distributes asymmetrically in early stages neurons A spatially localized Wnt3a source determines axon positioning and early guidance Concentration gradient of Wnt3a guides axonal outgrowth across a microfluidic chamber Wnt3a directly controls microtubules remodeling
Collapse
Affiliation(s)
- Eliana Stanganello
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Eitan Erez Zahavi
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Mithila Burute
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Jasper Smits
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Ingrid Jordens
- Oncode Institute, Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Madelon M Maurice
- Oncode Institute, Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lukas C Kapitein
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
24
|
Microtubule plus-end tracking Adenopolyposis Coli negatively regulates proplatelet formation. Sci Rep 2018; 8:15808. [PMID: 30361531 PMCID: PMC6202313 DOI: 10.1038/s41598-018-34118-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 10/03/2018] [Indexed: 12/17/2022] Open
Abstract
Platelets are produced upon profound reorganization of mature megakaryocytes (MK) leading to proplatelet elongation and release into the blood stream, a process termed thrombopoiesis. This highly dynamic process requires microtubules (MT) reorganization by mechanisms that are still incompletely understood. Adenomatous polyposis coli (APC) is a microtubule plus-end tracking protein involved in the regulation of MT in a number of cell systems and its inactivation has been reported to alter hematopoiesis. The aim of our study was to investigate the role of APC in megakaryopoiesis and the final steps of platelet formation. Down-regulation of APC in cultured human MK by RNA interference increased endomitosis and the proportion of cells able to extend proplatelets (68.8% (shAPC1) and 52.5% (shAPC2) vs 28.1% in the control). Similarly an increased ploidy and amplification of the proplatelet network were observed in MK differentiated from Lin- cells of mice with APC-deficiency in the MK lineage. In accordance, these mice exhibited increased platelet counts when compared to wild type mice (1,323 ± 111 vs 919 ± 52 platelets/µL; n = 12 p 0.0033**). Their platelets had a normal size, ultrastructure and number of microtubules coils and their main functions were also preserved. Loss of APC resulted in lower levels of acetylated tubulin and decreased activation of the Wnt signaling pathway. Thus, APC appears as an important regulator of proplatelet formation and overall thrombopoiesis.
Collapse
|
25
|
Jin T, Peng G, Wu E, Mendiratta S, Elul T. N-terminal and central domains of APC function to regulate branch number, length and angle in developing optic axonal arbors in vivo. Brain Res 2018; 1697:34-44. [PMID: 29856981 DOI: 10.1016/j.brainres.2018.05.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 05/07/2018] [Accepted: 05/28/2018] [Indexed: 11/24/2022]
Abstract
During formation of neuronal circuits, axons navigate long distances to reach their target locations in the brain. When axons arrive at their target tissues, in many cases, they extend collateral branches and/or terminal arbors that serve to increase the number of synaptic connections they make with target neurons. Here, we investigated how Adenomatous Polyposis Coli (APC) regulates terminal arborization of optic axons in living Xenopus laevis tadpoles. The N-terminal and central domains of APC that regulate the microtubule cytoskeleton and stability of β-catenin in the Wnt pathway, were co-expressed with GFP in individual optic axons, and their terminal arbors were then imaged in tectal midbrains of intact tadpoles. Our data show that the APCNTERM and APCβ-cat domains both decreased the mean number, and increased the mean length, of branches in optic axonal arbors relative to control arbors in vivo. Additional analysis demonstrated that expression of the APCNTERM domain increased the average bifurcation angle of branching in optic axonal arbors. However, the APCβ-cat domain did not significantly affect the mean branch angle of arbors in tecta of living tadpoles. These data suggest that APC N-terminal and central domains both modulate number and mean length of branches optic axonal arbors in a compensatory manner, but also define a specific function for the N-terminal domain of APC in regulating branch angle in optic axonal arbors in vivo. Our findings establish novel mechanisms for the multifunctional protein APC in shaping terminal arbors in the visual circuit of the developing vertebrate brain.
Collapse
Affiliation(s)
- Taegun Jin
- Touro University California, Vallejo, CA, United States
| | - Gregory Peng
- Touro University California, Vallejo, CA, United States
| | - Esther Wu
- Touro University California, Vallejo, CA, United States
| | | | - Tamira Elul
- Touro University California, Vallejo, CA, United States.
| |
Collapse
|
26
|
Expression and Manipulation of the APC-β-Catenin Pathway During Peripheral Neuron Regeneration. Sci Rep 2018; 8:13197. [PMID: 30181617 PMCID: PMC6123411 DOI: 10.1038/s41598-018-31167-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/03/2018] [Indexed: 02/07/2023] Open
Abstract
Molecules and pathways that suppress growth are expressed in postmitotic neurons, a potential advantage in mature neural networks, but a liability during regeneration. In this work, we probed the APC (adenomatous polyposis coli)-β-catenin partner pathway in adult peripheral sensory neurons during regeneration. APC had robust expression in the cytoplasm and perinuclear region of adult DRG sensory neurons both before and after axotomy injury. β-catenin was expressed in neuronal nuclei, neuronal cytoplasm and also in perineuronal satellite cells. In injured dorsal root ganglia (DRG) sensory neurons and their axons, we observed paradoxical APC upregulation, despite its role as an inhibitor of growth whereas β-catenin was downregulated. Inhibition of APC in adult sensory neurons and activation of β-catenin, LEF/TCF transcriptional factors were associated with increased neuronal plasticity in vitro. Local knockdown of APC, at the site of sciatic nerve crush injury enhanced evidence for electrophysiological, behavioural and structural regeneration in vivo. This was accompanied by upregulation of β-catenin. Collectively, the APC-β-catenin-LEF/TCF transcriptional pathway impacts intrinsic mechanisms of axonal regeneration and neuronal plasticity after injury, offering new options for addressing axon regeneration.
Collapse
|
27
|
Chronic infusion of Wnt7a, Wnt5a and Dkk-1 in the adult hippocampus induces structural synaptic changes and modifies anxiety and memory performance. Brain Res Bull 2018; 139:243-255. [DOI: 10.1016/j.brainresbull.2018.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/02/2018] [Accepted: 03/09/2018] [Indexed: 01/24/2023]
|
28
|
Arbeille E, Bashaw GJ. Brain Tumor promotes axon growth across the midline through interactions with the microtubule stabilizing protein Apc2. PLoS Genet 2018; 14:e1007314. [PMID: 29617376 PMCID: PMC5902039 DOI: 10.1371/journal.pgen.1007314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/16/2018] [Accepted: 03/19/2018] [Indexed: 11/20/2022] Open
Abstract
Commissural axons must cross the midline to establish reciprocal connections between the two sides of the body. This process is highly conserved between invertebrates and vertebrates and depends on guidance cues and their receptors to instruct axon trajectories. The DCC family receptor Frazzled (Fra) signals chemoattraction and promotes midline crossing in response to its ligand Netrin. However, in Netrin or fra mutants, the loss of crossing is incomplete, suggesting the existence of additional pathways. Here, we identify Brain Tumor (Brat), a tripartite motif protein, as a new regulator of midline crossing in the Drosophila CNS. Genetic analysis indicates that Brat acts independently of the Netrin/Fra pathway. In addition, we show that through its B-Box domains, Brat acts cell autonomously to regulate the expression and localization of Adenomatous polyposis coli-2 (Apc2), a key component of the Wnt canonical signaling pathway, to promote axon growth across the midline. Genetic evidence indicates that the role of Brat and Apc2 to promote axon growth across the midline is independent of Wnt and Beta-catenin-mediated transcriptional regulation. Instead, we propose that Brat promotes midline crossing through directing the localization or stability of Apc2 at the plus ends of microtubules in navigating commissural axons. These findings define a new mechanism in the coordination of axon growth and guidance at the midline. The establishment of neuronal connections that cross the midline of the animal is essential to generate neural circuits that coordinate the left and right sides of the body. Axons that cross the midline to form these connections are called commissural axons and the molecules and mechanisms that control midline axon crossing are remarkably conserved across animal evolution. In this study we have used a genetic screen in the fruit fly in an attempt to uncover additional players in this key developmental process, and have identified a novel role for the Brain Tumor (Brat) protein in promoting commissural axon growth across the midline. Unlike its previous described functions, in the context of midline axon guidance Brat cooperates with the microtubule stabilizing protein Apc2 to coordinate axon growth and guidance. Molecular and genetic analyses point to the conserved B box motifs of the Brat protein as key in promoting the association of Apc2 with the plus ends of microtubules. Brat is highly conserved and future studies will determine whether homologous genes play analogous roles in mammalian neural development.
Collapse
Affiliation(s)
- Elise Arbeille
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Greg J. Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
29
|
Liao CP, Li H, Lee HH, Chien CT, Pan CL. Cell-Autonomous Regulation of Dendrite Self-Avoidance by the Wnt Secretory Factor MIG-14/Wntless. Neuron 2018; 98:320-334.e6. [DOI: 10.1016/j.neuron.2018.03.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/06/2018] [Accepted: 03/16/2018] [Indexed: 11/26/2022]
|
30
|
Fassier C, Fréal A, Gasmi L, Delphin C, Ten Martin D, De Gois S, Tambalo M, Bosc C, Mailly P, Revenu C, Peris L, Bolte S, Schneider-Maunoury S, Houart C, Nothias F, Larcher JC, Andrieux A, Hazan J. Motor axon navigation relies on Fidgetin-like 1-driven microtubule plus end dynamics. J Cell Biol 2018. [PMID: 29535193 PMCID: PMC5940295 DOI: 10.1083/jcb.201604108] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Fassier et al. identify Fidgetin-like 1 (Fignl1) as a key growth cone (GC)-enriched microtubule (MT)-associated protein in motor circuit wiring. They show that Fignl1 modulates motor GC morphology and steering behavior by down-regulating EB binding at MT plus ends and promoting MT depolymerization beneath the cell cortex. During neural circuit assembly, extrinsic signals are integrated into changes in growth cone (GC) cytoskeleton underlying axon guidance decisions. Microtubules (MTs) were shown to play an instructive role in GC steering. However, the numerous actors required for MT remodeling during axon navigation and their precise mode of action are far from being deciphered. Using loss- and gain-of-function analyses during zebrafish development, we identify in this study the meiotic clade adenosine triphosphatase Fidgetin-like 1 (Fignl1) as a key GC-enriched MT-interacting protein in motor circuit wiring and larval locomotion. We show that Fignl1 controls GC morphology and behavior at intermediate targets by regulating MT plus end dynamics and growth directionality. We further reveal that alternative translation of Fignl1 transcript is a sophisticated mechanism modulating MT dynamics: a full-length isoform regulates MT plus end–tracking protein binding at plus ends, whereas shorter isoforms promote their depolymerization beneath the cell cortex. Our study thus pinpoints Fignl1 as a multifaceted key player in MT remodeling underlying motor circuit connectivity.
Collapse
Affiliation(s)
- Coralie Fassier
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Amélie Fréal
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Laïla Gasmi
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Christian Delphin
- Institut National de la Santé et de la Recherche Médicale U1216, Université Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France
| | - Daniel Ten Martin
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Stéphanie De Gois
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Monica Tambalo
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Christophe Bosc
- Institut National de la Santé et de la Recherche Médicale U1216, Université Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France
| | - Philippe Mailly
- Centre for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Céline Revenu
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
| | - Leticia Peris
- Institut National de la Santé et de la Recherche Médicale U1216, Université Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France
| | - Susanne Bolte
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Centre National de la Recherche Scientifique FR3631, Paris, France
| | - Sylvie Schneider-Maunoury
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Biologie du Développement, Centre National de la Recherche Scientifique UMR7622, Paris, France
| | - Corinne Houart
- Medical Research Council Centre for Developmental Neurobiology, King's College London, Guy's Hospital Campus, London, England, UK
| | - Fatiha Nothias
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Jean-Christophe Larcher
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Biologie du Développement, Centre National de la Recherche Scientifique UMR7622, Paris, France
| | - Annie Andrieux
- Institut National de la Santé et de la Recherche Médicale U1216, Université Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France
| | - Jamilé Hazan
- Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, Institut de Biologie Paris-Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| |
Collapse
|
31
|
Huang H, Yang T, Shao Q, Majumder T, Mell K, Liu G. Human TUBB3 Mutations Disrupt Netrin Attractive Signaling. Neuroscience 2018; 374:155-171. [PMID: 29382549 DOI: 10.1016/j.neuroscience.2018.01.046] [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: 10/01/2017] [Revised: 01/16/2018] [Accepted: 01/21/2018] [Indexed: 02/07/2023]
Abstract
Heterozygous missense mutations in human TUBB3 gene result in a spectrum of brain malformations associated with defects in axon guidance, neuronal migration and differentiation. However, the molecular mechanisms underlying mutation-related axon guidance abnormalities are unclear. Recent studies have shown that netrin-1, a canonical guidance cue, induced the interaction of TUBB3 with the netrin receptor deleted in colorectal cancer (DCC). Furthermore, TUBB3 is required for netrin-1-induced axon outgrowth, branching and pathfinding. Here, we provide evidence that TUBB3 mutations impair netrin/DCC signaling in the developing nervous system. The interaction of DCC with most TUBB3 mutants (eight out of twelve) is significantly reduced compared to the wild-type TUBB3. TUBB3 mutants R262C and A302V exhibit decreased subcellular colocalization with DCC in the growth cones of primary neurons. Netrin-1 increases the interaction of endogenous DCC with wild-type human TUBB3, but not R262C or A302V, in primary neurons. Netrin-1 also increases co-sedimentation of DCC with polymerized microtubules (MTs) in primary neurons expressing the wild-type TUBB3, but not R262C or A302V. Expression of either R262C or A302V not only suppresses netrin-1-induced neurite outgrowth, branching and attraction in vitro, but also causes defects in spinal cord commissural axon (CA) projection and pathfinding in ovo. Our study reveals that missense TUBB3 mutations specifically disrupt netrin/DCC-mediated attractive signaling.
Collapse
Affiliation(s)
- Huai Huang
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Kristopher Mell
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA.
| |
Collapse
|
32
|
Garcia AL, Udeh A, Kalahasty K, Hackam AS. A growing field: The regulation of axonal regeneration by Wnt signaling. Neural Regen Res 2018; 13:43-52. [PMID: 29451203 PMCID: PMC5840987 DOI: 10.4103/1673-5374.224359] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The canonical Wnt/β-catenin pathway is a highly conserved signaling cascade that plays critical roles during embryogenesis. Wnt ligands regulate axonal extension, growth cone guidance and synaptogenesis throughout the developing central nervous system (CNS). Recently, studies in mammalian and fish model systems have demonstrated that Wnt/β-catenin signaling also promotes axonal regeneration in the adult optic nerve and spinal cord after injury, raising the possibility that Wnt could be developed as a therapeutic strategy. In this review, we summarize experimental evidence that reveals novel roles for Wnt signaling in the injured CNS, and discuss possible mechanisms by which Wnt ligands could overcome molecular barriers inhibiting axonal growth to promote regeneration. A central challenge in the neuroscience field is developing therapeutic strategies that induce robust axonal regeneration. Although adult axons have the capacity to respond to axonal guidance molecules after injury, there are several major obstacles for axonal growth, including extensive neuronal death, glial scars at the injury site, and lack of axonal guidance signals. Research in rodents demonstrated that activation of Wnt/β-catenin signaling in retinal neurons and radial glia induced neuronal survival and axonal growth, but that activation within reactive glia at the injury site promoted proliferation and glial scar formation. Studies in zebrafish spinal cord injury models confirm an axonal regenerative role for Wnt/β-catenin signaling and identified the cell types responsible. Additionally, in vitro and in vivo studies demonstrated that Wnt induces axonal and neurite growth through transcription-dependent effects of its central mediator β-catenin, potentially by inducing regeneration-promoting genes. Canonical Wnt signaling may also function through transcription-independent interactions of β-catenin with cytoskeletal elements, which could stabilize growing axons and control growth cone movement. Therefore, these studies suggest that Wnt-induced pathways responsible for regulating axonal growth during embryogenesis could be repurposed to promote axonal growth after injury.
Collapse
Affiliation(s)
- Armando L Garcia
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Adanna Udeh
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Karthik Kalahasty
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abigail S Hackam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
33
|
Zwamborn RA, Snijders C, An N, Thomson A, Rutten BP, de Nijs L. Wnt Signaling in the Hippocampus in Relation to Neurogenesis, Neuroplasticity, Stress and Epigenetics. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 158:129-157. [DOI: 10.1016/bs.pmbts.2018.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
34
|
Zheng C, Diaz-Cuadros M, Nguyen KCQ, Hall DH, Chalfie M. Distinct effects of tubulin isotype mutations on neurite growth in Caenorhabditis elegans. Mol Biol Cell 2017; 28:2786-2801. [PMID: 28835377 PMCID: PMC5638583 DOI: 10.1091/mbc.e17-06-0424] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022] Open
Abstract
Different tubulin isotypes perform different functions in the regulation of microtubule (MT) structure and neurite growth, and missense mutations of tubulin genes have three types of distinct effects on MT stability and neurite growth. One α-tubulin isotype appears to induce relative instability due to the lack of potential posttranslational modification sites. Tubulins, the building block of microtubules (MTs), play a critical role in both supporting and regulating neurite growth. Eukaryotic genomes contain multiple tubulin isotypes, and their missense mutations cause a range of neurodevelopmental defects. Using the Caenorhabditis elegans touch receptor neurons, we analyzed the effects of 67 tubulin missense mutations on neurite growth. Three types of mutations emerged: 1) loss-of-function mutations, which cause mild defects in neurite growth; 2) antimorphic mutations, which map to the GTP binding site and intradimer and interdimer interfaces, significantly reduce MT stability, and cause severe neurite growth defects; and 3) neomorphic mutations, which map to the exterior surface, increase MT stability, and cause ectopic neurite growth. Structure-function analysis reveals a causal relationship between tubulin structure and MT stability. This stability affects neuronal morphogenesis. As part of this analysis, we engineered several disease-associated human tubulin mutations into C. elegans genes and examined their impact on neuronal development at the cellular level. We also discovered an α-tubulin (TBA-7) that appears to destabilize MTs. Loss of TBA-7 led to the formation of hyperstable MTs and the generation of ectopic neurites; the lack of potential sites for polyamination and polyglutamination on TBA-7 may be responsible for this destabilization.
Collapse
Affiliation(s)
- Chaogu Zheng
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | | | - Ken C Q Nguyen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - David H Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Martin Chalfie
- Department of Biological Sciences, Columbia University, New York, NY 10027
| |
Collapse
|
35
|
Mustyatsa VV, Boyakhchyan AV, Ataullakhanov FI, Gudimchuk NB. EB-family proteins: Functions and microtubule interaction mechanisms. BIOCHEMISTRY (MOSCOW) 2017; 82:791-802. [DOI: 10.1134/s0006297917070045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Armijo-Weingart L, Gallo G. It takes a village to raise a branch: Cellular mechanisms of the initiation of axon collateral branches. Mol Cell Neurosci 2017; 84:36-47. [PMID: 28359843 DOI: 10.1016/j.mcn.2017.03.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/03/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022] Open
Abstract
The formation of axon collateral branches from the pre-existing shafts of axons is an important aspect of neurodevelopment and the response of the nervous system to injury. This article provides an overview of the role of the cytoskeleton and signaling mechanisms in the formation of axon collateral branches. Both the actin filament and microtubule components of the cytoskeleton are required for the formation of axon branches. Recent work has begun to shed light on how these two elements of the cytoskeleton are integrated by proteins that functionally or physically link the cytoskeleton. While a number of signaling pathways have been determined as having a role in the formation of axon branches, the complexity of the downstream mechanisms and links to specific signaling pathways remain to be fully determined. The regulation of intra-axonal protein synthesis and organelle function are also emerging as components of signal-induced axon branching. Although much has been learned in the last couple of decades about the mechanistic basis of axon branching we can look forward to continue elucidating this complex biological phenomenon with the aim of understanding how multiple signaling pathways, cytoskeletal regulators and organelles are coordinated locally along the axon to give rise to a branch.
Collapse
Affiliation(s)
- Lorena Armijo-Weingart
- Shriners Pediatric Research Center, Temple University, Department of Anatomy and Cell Biology, 3500 North Broad St, Philadelphia, PA 19140, United States
| | - Gianluca Gallo
- Shriners Pediatric Research Center, Temple University, Department of Anatomy and Cell Biology, 3500 North Broad St, Philadelphia, PA 19140, United States.
| |
Collapse
|
37
|
Mulligan KA, Cheyette BNR. Neurodevelopmental Perspectives on Wnt Signaling in Psychiatry. MOLECULAR NEUROPSYCHIATRY 2017; 2:219-246. [PMID: 28277568 DOI: 10.1159/000453266] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mounting evidence indicates that Wnt signaling is relevant to pathophysiology of diverse mental illnesses including schizophrenia, bipolar disorder, and autism spectrum disorder. In the 35 years since Wnt ligands were first described, animal studies have richly explored how downstream Wnt signaling pathways affect an array of neurodevelopmental processes and how their disruption can lead to both neurological and behavioral phenotypes. Recently, human induced pluripotent stem cell (hiPSC) models have begun to contribute to this literature while pushing it in increasingly translational directions. Simultaneously, large-scale human genomic studies are providing evidence that sequence variation in Wnt signal pathway genes contributes to pathogenesis in several psychiatric disorders. This article reviews neurodevelopmental and postneurodevelopmental functions of Wnt signaling, highlighting mechanisms, whereby its disruption might contribute to psychiatric illness, and then reviews the most reliable recent genetic evidence supporting that mutations in Wnt pathway genes contribute to psychiatric illness. We are proponents of the notion that studies in animal and hiPSC models informed by the human genetic data combined with the deep knowledge base and tool kits generated over the last several decades of basic neurodevelopmental research will yield near-term tangible advances in neuropsychiatry.
Collapse
Affiliation(s)
- Kimberly A Mulligan
- Department of Biological Sciences, California State University, Sacramento, CA, USA
| | - Benjamin N R Cheyette
- Department of Psychiatry, Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
38
|
Chen T, Yu Y, Tang LJ, Kong L, Zhang CH, Chu HY, Yin LW, Ma HY. Neural stem cells over-expressing brain-derived neurotrophic factor promote neuronal survival and cytoskeletal protein expression in traumatic brain injury sites. Neural Regen Res 2017; 12:433-439. [PMID: 28469658 PMCID: PMC5399721 DOI: 10.4103/1673-5374.202947] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Cytoskeletal proteins are involved in neuronal survival. Brain-derived neurotrophic factor can increase expression of cytoskeletal proteins during regeneration after axonal injury. However, the effect of neural stem cells genetically modified by brain-derived neurotrophic factor transplantation on neuronal survival in the injury site still remains unclear. To examine this, we established a rat model of traumatic brain injury by controlled cortical impact. At 72 hours after injury, 2 × 107 cells/mL neural stem cells overexpressing brain-derived neurotrophic factor or naive neural stem cells (3 mL) were injected into the injured cortex. At 1–3 weeks after transplantation, expression of neurofilament 200, microtubule-associated protein 2, actin, calmodulin, and beta-catenin were remarkably increased in the injury sites. These findings confirm that brain-derived neurotrophic factor-transfected neural stem cells contribute to neuronal survival, growth, and differentiation in the injury sites. The underlying mechanisms may be associated with increased expression of cytoskeletal proteins and the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yan Yu
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Liu-Jiu Tang
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Li Kong
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Cheng-Hong Zhang
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Hai-Ying Chu
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Liang-Wei Yin
- Department of Oncology, Dalian Central Hospital, Dalian, Liaoning Province, China
| | - Hai-Ying Ma
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
39
|
Patel AK, Park KK, Hackam AS. Wnt signaling promotes axonal regeneration following optic nerve injury in the mouse. Neuroscience 2016; 343:372-383. [PMID: 28011153 DOI: 10.1016/j.neuroscience.2016.12.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 12/07/2016] [Accepted: 12/12/2016] [Indexed: 02/08/2023]
Abstract
Adult mammalian CNS axons generally do not regenerate, creating an obstacle to effective repair and recovery after neuronal injury. The canonical Wnt/β-catenin signaling pathway is an essential signal transduction cascade that regulates axon growth and neurite extension in the developing mammalian embryo. In this study, we investigated whether a Wnt/β-catenin signaling activator could be repurposed to induce regeneration in the adult CNS after axonal injury. We used a retinal ganglion cell (RGC) axon crush injury model in a transgenic Wnt reporter mouse, and intravitreal injections were used to deliver Wnt3a or saline to the RGC cell bodies within the retina. Our findings demonstrated that Wnt3a induced Wnt signaling in RGCs and resulted in significant axonal regrowth past the lesion site when measured at two and four weeks post-injury. Furthermore, Wnt3a-injected eyes showed increased survival of RGCs and significantly higher pattern electroretinography (PERG) amplitudes compared to the control. Additionally, Wnt3a-induced axonal regeneration and RGC survival were associated with elevated activation of the transcription factor Stat3, and reducing expression of Stat3 using a conditional Stat3 knock-out mouse line led to diminished Wnt3a-dependent axonal regeneration and RGC survival. Therefore, these findings reveal a novel role for retinal Wnt signaling in axonal regrowth and RGC survival following axonal injury, which may lead to the development of novel therapies for axonal regeneration.
Collapse
Affiliation(s)
- Amit K Patel
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Kevin K Park
- Miami Project to Cure Paralysis, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Abigail S Hackam
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
40
|
Marzo A, Galli S, Lopes D, McLeod F, Podpolny M, Segovia-Roldan M, Ciani L, Purro S, Cacucci F, Gibb A, Salinas PC. Reversal of Synapse Degeneration by Restoring Wnt Signaling in the Adult Hippocampus. Curr Biol 2016; 26:2551-2561. [PMID: 27593374 PMCID: PMC5070786 DOI: 10.1016/j.cub.2016.07.024] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/05/2016] [Accepted: 07/12/2016] [Indexed: 11/30/2022]
Abstract
Synapse degeneration occurs early in neurodegenerative diseases and correlates strongly with cognitive decline in Alzheimer’s disease (AD). The molecular mechanisms that trigger synapse vulnerability and those that promote synapse regeneration after substantial synaptic failure remain poorly understood. Increasing evidence suggests a link between a deficiency in Wnt signaling and AD. The secreted Wnt antagonist Dickkopf-1 (Dkk1), which is elevated in AD, contributes to amyloid-β-mediated synaptic failure. However, the impact of Dkk1 at the circuit level and the mechanism by which synapses disassemble have not yet been explored. Using a transgenic mouse model that inducibly expresses Dkk1 in the hippocampus, we demonstrate that Dkk1 triggers synapse loss, impairs long-term potentiation, enhances long-term depression, and induces learning and memory deficits. We decipher the mechanism involved in synapse loss induced by Dkk1 as it can be prevented by combined inhibition of the Gsk3 and RhoA-Rock pathways. Notably, after loss of synaptic connectivity, reactivation of the Wnt pathway by cessation of Dkk1 expression completely restores synapse number, synaptic plasticity, and long-term memory. These findings demonstrate the remarkable capacity of adult neurons to regenerate functional circuits and highlight Wnt signaling as a targetable pathway for neuronal circuit recovery after synapse degeneration. Wnt signaling is required for synapse integrity in the adult hippocampus Dkk1 induces synapse loss and deficits in synaptic plasticity and long-term memory Dkk1 disassembles synapses by activating the Gsk3 and Rock pathways Synapse loss and memory defects are reversible by reactivation of the Wnt pathway
Collapse
Affiliation(s)
- Aude Marzo
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Soledad Galli
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Douglas Lopes
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Faye McLeod
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Marina Podpolny
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | | | - Lorenza Ciani
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Silvia Purro
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Francesca Cacucci
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, UK
| | - Alasdair Gibb
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, UK
| | - Patricia C Salinas
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
41
|
Tumour Suppressor Adenomatous Polyposis Coli (APC) localisation is regulated by both Kinesin-1 and Kinesin-2. Sci Rep 2016; 6:27456. [PMID: 27272132 PMCID: PMC4895226 DOI: 10.1038/srep27456] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/17/2016] [Indexed: 12/18/2022] Open
Abstract
Microtubules and their associated proteins (MAPs) underpin the polarity of specialised cells. Adenomatous polyposis coli (APC) is one such MAP with a multifunctional agenda that requires precise intracellular localisations. Although APC has been found to associate with kinesin-2 subfamily members, the exact mechanism for the peripheral localization of APC remains unclear. Here we show that the heavy chain of kinesin-1 directly interacts with the APC C-terminus, contributing to the peripheral localisation of APC in fibroblasts. In rat hippocampal neurons the kinesin-1 binding domain of APC is required for its axon tip enrichment. Moreover, we demonstrate that APC requires interactions with both kinesin-2 and kinesin-1 for this localisation. Underlining the importance of the kinesin-1 association, neurons expressing APC lacking kinesin-1-binding domain have shorter axons. The identification of this novel kinesin-1-APC interaction highlights the complexity and significance of APC localisation in neurons.
Collapse
|
42
|
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: 63] [Impact Index Per Article: 7.0] [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
|
43
|
Cammarata GM, Bearce EA, Lowery LA. Cytoskeletal social networking in the growth cone: How +TIPs mediate microtubule-actin cross-linking to drive axon outgrowth and guidance. Cytoskeleton (Hoboken) 2016; 73:461-76. [PMID: 26783725 DOI: 10.1002/cm.21272] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/08/2016] [Accepted: 01/08/2016] [Indexed: 11/08/2022]
Abstract
The growth cone is a unique structure capable of guiding axons to their proper destinations. Within the growth cone, extracellular guidance cues are interpreted and then transduced into physical changes in the actin filament (F-actin) and microtubule cytoskeletons, providing direction and movement. While both cytoskeletal networks individually possess important growth cone-specific functions, recent data over the past several years point towards a more cooperative role between the two systems. Facilitating this interaction between F-actin and microtubules, microtubule plus-end tracking proteins (+TIPs) have been shown to link the two cytoskeletons together. Evidence suggests that many +TIPs can couple microtubules to F-actin dynamics, supporting both microtubule advance and retraction in the growth cone periphery. In addition, growing in vitro and in vivo data support a secondary role for +TIPs in which they may participate as F-actin nucleators, thus directly influencing F-actin dynamics and organization. This review focuses on how +TIPs may link F-actin and microtubules together in the growth cone, and how these interactions may influence axon guidance. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Laura Anne Lowery
- Department of Biology, Boston College, Chestnut Hill, Massachusetts.
| |
Collapse
|
44
|
Michalski JP, Cummings SE, O'Meara RW, Kothary R. Integrin-linked kinase regulates oligodendrocyte cytoskeleton, growth cone, and adhesion dynamics. J Neurochem 2016; 136:536-49. [DOI: 10.1111/jnc.13446] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/15/2015] [Accepted: 11/20/2015] [Indexed: 01/28/2023]
Affiliation(s)
- John-Paul Michalski
- Ottawa Hospital Research Institute; Ottawa Ontario Canada
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa Ontario Canada
| | - Sarah E. Cummings
- Ottawa Hospital Research Institute; Ottawa Ontario Canada
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa Ontario Canada
| | - Ryan W. O'Meara
- Ottawa Hospital Research Institute; Ottawa Ontario Canada
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa Ontario Canada
| | - Rashmi Kothary
- Ottawa Hospital Research Institute; Ottawa Ontario Canada
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa Ontario Canada
- Department of Medicine; University of Ottawa; Ottawa Ontario Canada
- University of Ottawa Centre for Neuromuscular Disease; Ottawa Ontario Canada
| |
Collapse
|
45
|
Coullery RP, Ferrari ME, Rosso SB. Neuronal development and axon growth are altered by glyphosate through a WNT non-canonical signaling pathway. Neurotoxicology 2016; 52:150-61. [PMID: 26688330 DOI: 10.1016/j.neuro.2015.12.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 01/25/2023]
Abstract
The growth and morphological differentiation of neurons are critical events in the establishment of proper neuronal connectivity and functioning. The developing nervous system is highly susceptible to damage caused by exposure to environmental contaminants. Glyphosate-containing herbicides are the most used agrochemicals in the world, particularly on genetically modified plants. Previous studies have demonstrated that glyphosate induces neurotoxicity in mammals. Therefore, its action mechanism on the nervous system needs to be determined. In this study, we report about impaired neuronal development caused by glyphosate exposure. Particularly, we observed that the initial axonal differentiation and growth of cultured neurons is affected by glyphosate since most treated cells remained undifferentiated after 1 day in culture. Although they polarized at 2 days in vitro, they elicited shorter and unbranched axons and they also developed less complex dendritic arbors compared to controls. To go further, we attempted to identify the cellular mechanism by which glyphosate affected neuronal morphology. Biochemical approaches revealed that glyphosate led to a decrease in Wnt5a level, a key factor for the initial neurite development and maturation, as well as inducing a down-regulation of CaMKII activity. This data suggests that the morphological defects would likely be a consequence of the decrease in both Wnt5a expression and CaMKII activity induced by glyphosate. Additionally, these changes might be reflected in a subsequent neuronal dysfunction. Therefore, our findings highlight the importance of establishing rigorous control on the use of glyphosate-based herbicides in order to protect mammals' health.
Collapse
Affiliation(s)
- Romina P Coullery
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - María E Ferrari
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Silvana B Rosso
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina.
| |
Collapse
|
46
|
Selvaraj P, Huang JSW, Chen A, Skalka N, Rosin-Arbesfeld R, Loh YP. Neurotrophic factor-α1 modulates NGF-induced neurite outgrowth through interaction with Wnt-3a and Wnt-5a in PC12 cells and cortical neurons. Mol Cell Neurosci 2015; 68:222-33. [PMID: 26276171 DOI: 10.1016/j.mcn.2015.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/22/2015] [Accepted: 08/03/2015] [Indexed: 02/06/2023] Open
Abstract
Wnt-3a and Wnt-5a signaling activities inhibit and promote neurite outgrowth, respectively, to regulate dendritic and axonal genesis during neurodevelopment. NF-α1, a neurotrophic factor, has been shown to modulate dendritic remodeling and negatively regulate the canonical Wnt-3a pathway. Here, we investigated whether NF-α1 could modify nerve growth factor (NGF)-induced neurite outgrowth through interaction with Wnt-3a and Wnt-5a in PC12 cells and mouse primary cortical neurons. We showed that NGF-induced neurite outgrowth was inhibited by Wnt-3a, and this inhibition was prevented by NF-α1. Western blot analysis revealed that NF-α1 reduced the expression of both β-catenin in the canonical Wnt-3a pathway and Rho, a downstream effector of Wnt-3a's non-canonical signaling pathway. Treatment of PC12 cells with a ROCK inhibitor prevented the inhibition of NGF-induced neurite outgrowth by Wnt-3a, suggesting that NF-α1 promotes neurite outgrowth in the presence of Wnt-3a by down-regulating its canonical and non-canonical activities. Interestingly, treatment of PC12 cells with Wnt-5a, which formed a complex with NF-α1, induced neurite outgrowth that was enhanced by treatment with the combination of Wnt-5a, NGF, and NF-α1. These effects of NF-α1 on Wnt 3a's and Wnt 5a's regulation of neurite outgrowth in PC12 cells were also demonstrated in primary cultures of mouse cortical neurons. In addition, we showed in PC12 cells that NF-α1 acts by upregulating adenomatous polyposis coli (APC) accumulation at neurite tips, thereby providing positive and negative Wnt-3a/Wnt-5a mediated cues to modulate neurite outgrowth, a process important during neurodevelopment.
Collapse
Affiliation(s)
- Prabhuanand Selvaraj
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Jane S W Huang
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Alexander Chen
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Nir Skalka
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rina Rosin-Arbesfeld
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Y Peng Loh
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
47
|
Stamatakou E, Hoyos-Flight M, Salinas PC. Wnt Signalling Promotes Actin Dynamics during Axon Remodelling through the Actin-Binding Protein Eps8. PLoS One 2015; 10:e0134976. [PMID: 26252776 PMCID: PMC4529215 DOI: 10.1371/journal.pone.0134976] [Citation(s) in RCA: 14] [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: 01/15/2015] [Accepted: 07/07/2015] [Indexed: 11/26/2022] Open
Abstract
Upon arrival at their synaptic targets, axons slow down their growth and extensively remodel before the assembly of presynaptic boutons. Wnt proteins are target-derived secreted factors that promote axonal remodelling and synaptic assembly. In the developing spinal cord, Wnts secreted by motor neurons promote axonal remodelling of NT-3 responsive dorsal root ganglia neurons. Axon remodelling induced by Wnts is characterised by growth cone pausing and enlargement, processes that depend on the re-organisation of microtubules. However, the contribution of the actin cytoskeleton has remained unexplored. Here, we demonstrate that Wnt3a regulates the actin cytoskeleton by rapidly inducing F-actin accumulation in growth cones from rodent DRG neurons through the scaffold protein Dishevelled-1 (Dvl1) and the serine-threonine kinase Gsk3β. Importantly, these changes in actin cytoskeleton occurs before enlargement of the growth cones is evident. Time-lapse imaging shows that Wnt3a increases lamellar protrusion and filopodia velocity. In addition, pharmacological inhibition of actin assembly demonstrates that Wnt3a increases actin dynamics. Through a yeast-two hybrid screen, we identified the actin-binding protein Eps8 as a direct interactor of Dvl1, a scaffold protein crucial for the Wnt signalling pathway. Gain of function of Eps8 mimics Wnt-mediated axon remodelling, whereas Eps8 silencing blocks the axon remodelling activity of Wnt3a. Importantly, blockade of the Dvl1-Eps8 interaction completely abolishes Wnt3a-mediated axonal remodelling. These findings demonstrate a novel role for Wnt-Dvl1 signalling through Eps8 in the regulation of axonal remodeling.
Collapse
Affiliation(s)
- Eleanna Stamatakou
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Monica Hoyos-Flight
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Patricia C. Salinas
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
Bearce EA, Erdogan B, Lowery LA. TIPsy tour guides: how microtubule plus-end tracking proteins (+TIPs) facilitate axon guidance. Front Cell Neurosci 2015; 9:241. [PMID: 26175669 PMCID: PMC4485311 DOI: 10.3389/fncel.2015.00241] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/15/2015] [Indexed: 01/01/2023] Open
Abstract
The growth cone is a dynamic cytoskeletal vehicle, which drives the end of a developing axon. It serves to interpret and navigate through the complex landscape and guidance cues of the early nervous system. The growth cone’s distinctive cytoskeletal organization offers a fascinating platform to study how extracellular cues can be translated into mechanical outgrowth and turning behaviors. While many studies of cell motility highlight the importance of actin networks in signaling, adhesion, and propulsion, both seminal and emerging works in the field have highlighted a unique and necessary role for microtubules (MTs) in growth cone navigation. Here, we focus on the role of singular pioneer MTs, which extend into the growth cone periphery and are regulated by a diverse family of microtubule plus-end tracking proteins (+TIPs). These +TIPs accumulate at the dynamic ends of MTs, where they are well-positioned to encounter and respond to key signaling events downstream of guidance receptors, catalyzing immediate changes in microtubule stability and actin cross-talk, that facilitate both axonal outgrowth and turning events.
Collapse
Affiliation(s)
| | - Burcu Erdogan
- Department of Biology, Boston College Chestnut Hill, MA, USA
| | | |
Collapse
|
49
|
Sugie A, Hakeda-Suzuki S, Suzuki E, Silies M, Shimozono M, Möhl C, Suzuki T, Tavosanis G. Molecular Remodeling of the Presynaptic Active Zone of Drosophila Photoreceptors via Activity-Dependent Feedback. Neuron 2015; 86:711-25. [PMID: 25892303 DOI: 10.1016/j.neuron.2015.03.046] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 02/10/2015] [Accepted: 03/18/2015] [Indexed: 11/27/2022]
Abstract
Neural activity contributes to the regulation of the properties of synapses in sensory systems, allowing for adjustment to a changing environment. Little is known about how synaptic molecular components are regulated to achieve activity-dependent plasticity at central synapses. Here, we found that after prolonged exposure to natural ambient light the presynaptic active zone in Drosophila photoreceptors undergoes reversible remodeling, including loss of Bruchpilot, DLiprin-α, and DRBP, but not of DSyd-1 or Cacophony. The level of depolarization of the postsynaptic neurons is critical for the light-induced changes in active zone composition in the photoreceptors, indicating the existence of a feedback signal. In search of this signal, we have identified a crucial role of microtubule meshwork organization downstream of the divergent canonical Wnt pathway, potentially via Kinesin-3 Imac. These data reveal that active zone composition can be regulated in vivo and identify the underlying molecular machinery.
Collapse
Affiliation(s)
- Atsushi Sugie
- Dendrite Differentiation, German Center for Neurodegenerative Diseases (DZNE), Bonn 53175, Germany
| | - Satoko Hakeda-Suzuki
- Core Division of Advanced Research, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology (Titech), Yokohama 226-8501, Japan
| | - Emiko Suzuki
- Gene Network Laboratory, National Institute of Genetics and Department of Genetics, SOKENDAI, Mishima 411-8540, Japan
| | - Marion Silies
- European Neuroscience Institute (ENI), 37077 Göttingen, Germany
| | - Mai Shimozono
- Core Division of Advanced Research, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology (Titech), Yokohama 226-8501, Japan
| | - Christoph Möhl
- Image and Data Analysis Facility, German Center for Neurodegenerative Diseases (DZNE), Bonn 53175, Germany
| | - Takashi Suzuki
- Core Division of Advanced Research, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology (Titech), Yokohama 226-8501, Japan.
| | - Gaia Tavosanis
- Dendrite Differentiation, German Center for Neurodegenerative Diseases (DZNE), Bonn 53175, Germany.
| |
Collapse
|
50
|
Wnt signalling in neuronal differentiation and development. Cell Tissue Res 2014; 359:215-23. [PMID: 25234280 DOI: 10.1007/s00441-014-1996-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/25/2014] [Indexed: 12/15/2022]
Abstract
Wnts are secreted glycoproteins that play multiple roles in early development, including the differentiation of precursor cells. During this period, gradients of Wnts and other morphogens are formed and regulate the differentiation and migration of neural progenitor cells. Afterwards, Wnt signalling cascades participate in the formation of neuronal circuits, playing roles in dendrite and axon development, dendritic spine formation and synaptogenesis. Finally, in the adult brain, Wnts control hippocampal plasticity, regulating synaptic transmission and neurogenesis. In this review, we summarize the reported roles of Wnt signalling cascades in these processes with a particular emphasis on the role of Wnts in neuronal differentiation and development.
Collapse
|