1
|
Fréal A, Hoogenraad CC. The dynamic axon initial segment: From neuronal polarity to network homeostasis. Neuron 2025; 113:649-669. [PMID: 39947181 DOI: 10.1016/j.neuron.2025.01.004] [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: 04/25/2024] [Revised: 09/15/2024] [Accepted: 01/07/2025] [Indexed: 03/08/2025]
Abstract
The axon initial segment (AIS) is a highly specialized compartment in neurons that resides in between axonal and somatodendritic domains. The localization of the AIS in the proximal part of the axon is essential for its two major functions: generating and modulating action potentials and maintaining neuron polarity. Recent findings revealed that the incredibly stable AIS is generated from highly dynamic components and can undergo extensive structural and functional changes in response to alterations in activity levels. These activity-dependent alterations of AIS structure and function have profound consequences for neuronal functioning, and AIS plasticity has emerged as a key regulator of network homeostasis. This review highlights the functions of the AIS, its architecture, and how its organization and remodeling are influenced by developmental plasticity and both acute and chronic adaptations. It also discusses the mechanisms underlying these processes and explores how dysregulated AIS plasticity may contribute to brain disorders.
Collapse
Affiliation(s)
- Amélie Fréal
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam, the Netherlands
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech, Inc, South San Francisco, CA 94080, USA.
| |
Collapse
|
2
|
Heydemann L, Ciurkiewicz M, Störk T, Zdora I, Hülskötter K, Gregor KM, Michaely LM, Reineking W, Schreiner T, Beythien G, Volz A, Tuchel T, Meyer Zu Natrup C, Schünemann LM, Clever S, Henneck T, von Köckritz-Blickwede M, Schaudien D, Rohn K, Schughart K, Geffers R, Kaneko MK, Kato Y, Gross C, Amanakis G, Pavlou A, Baumgärtner W, Armando F. Respiratory long COVID in aged hamsters features impaired lung function post-exercise with bronchiolization and fibrosis. Nat Commun 2025; 16:2080. [PMID: 40021627 PMCID: PMC11871369 DOI: 10.1038/s41467-025-57267-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
Long-term consequences of SARS-CoV-2 infection affect millions of people and strain public health systems. The underlying pathomechanisms remain unclear, necessitating further research in appropriate animal models. This study aimed to characterize the trajectory of lung regeneration over 112 days in the male hamster model by combining morphological, transcriptomic and functional readouts. We demonstrate that in the acute phase, SARS-CoV-2 Delta-infected, male, aged hamsters show a severe impairment of lung function at rest. In the chronic phase, similar impairments persisted up to 7 weeks post-infection but were only evident after exercise on a rodent treadmill. The male hamster model recapitulates chronic pulmonary fibrotic changes observed in many patients with respiratory long COVID, but lacks extra-pulmonary long-term lesions. We show that sub-pleural and interstitial pulmonary fibrosis as well as alveolar bronchiolization persist until 112 dpi. Interestingly, CK8+ alveolar differentiation intermediate (ADI) cells are becoming less prominent in the alveolar proliferation areas from 28 dpi on. Instead, CK14+ airway basal cells and SCGB1A1+ club cells, expressing cell proliferation markers, mainly populate alveolar bronchiolization areas at later time-points. We postulate that pulmonary fibrosis and SCGB1A1+ club cell-rich areas of alveolar bronchiolization represent potential risk factors for other diseases in long-COVID survivors.
Collapse
Affiliation(s)
- Laura Heydemann
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | | | - Theresa Störk
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Isabel Zdora
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Kirsten Hülskötter
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | | | | | - Wencke Reineking
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Tom Schreiner
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Georg Beythien
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Asisa Volz
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Tamara Tuchel
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Christian Meyer Zu Natrup
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Lisa-Marie Schünemann
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Sabrina Clever
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Timo Henneck
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
- Department of Biochemistry, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Maren von Köckritz-Blickwede
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
- Department of Biochemistry, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Karl Rohn
- Department of Biometry, Epidemiology and Data Management, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Klaus Schughart
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Institute of Virology Münster, University of Münster, Münster, Germany
| | - Robert Geffers
- Helmholtz Centre for Infection Research (HZI), Brunswick, Germany
| | - Mika K Kaneko
- Department of antibody drug development, Tohoku University, Sendai, Miyagi, Japan
| | - Yukinari Kato
- Department of antibody drug development, Tohoku University, Sendai, Miyagi, Japan
| | - Carina Gross
- Department of Cardiology and Angiology, Hanover Medical School (MHH), Hanover, Germany
| | - Georgios Amanakis
- Department of Cardiology and Angiology, Hanover Medical School (MHH), Hanover, Germany
| | - Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany.
| | - Federico Armando
- Pathology Unit, Department of Veterinary Science, University of Parma, Parma, Italy
| |
Collapse
|
3
|
Behrens KA, Koblmüller S, Kocher TD. Genome assemblies for Chromidotilapia guntheri (Teleostei: Cichlidae) identify a novel candidate gene for vertebrate sex determination, RIN3. Front Genet 2024; 15:1447628. [PMID: 39221227 PMCID: PMC11361979 DOI: 10.3389/fgene.2024.1447628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Advances in genome sequencing have greatly accelerated the identification of sex chromosomes in a variety of species. Many of these species have experienced structural rearrangements that reduce recombination between the sex chromosomes, allowing the accumulation of sequence differences over many megabases. Identification of the genes that are responsible for sex determination within these sometimes large regions has proved difficult. Here, we identify an XY sex chromosome system on LG19 in the West African cichlid fish Chromidotilapia guntheri in which the region of differentiation extends over less than 400 kb. We develop high-quality male and female genome assemblies for this species, which confirm the absence of structural variants, and which facilitate the annotation of genes in the region. The peak of differentiation lies within rin3, which has experienced several debilitating mutations on the Y chromosome. We suggest two hypotheses about how these mutations might disrupt endocytosis, leading to Mendelian effects on sexual development.
Collapse
Affiliation(s)
- Kristen A. Behrens
- Department of Biology, University of Maryland, College Park, MD, United States
| | | | - Thomas D. Kocher
- Department of Biology, University of Maryland, College Park, MD, United States
| |
Collapse
|
4
|
Zehtabian A, Fuchs J, Eickholt BJ, Ewers H. Automated Analysis of Neuronal Morphology in 2D Fluorescence Micrographs through an Unsupervised Semantic Segmentation of Neurons. Neuroscience 2024; 551:333-344. [PMID: 38838980 DOI: 10.1016/j.neuroscience.2024.05.024] [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: 12/19/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024]
Abstract
Brain function emerges from a highly complex network of specialized cells that are interlinked by billions of synapses. The synaptic connectivity between neurons is established between the elongated processes of their axons and dendrites or, together, neurites. To establish these connections, cellular neurites have to grow in highly specialized, cell-type dependent patterns covering extensive distances and connecting with thousands of other neurons. The outgrowth and branching of neurites are tightly controlled during development and are a commonly used functional readout of imaging in the neurosciences. Manual analysis of neuronal morphology from microscopy images, however, is very time intensive and prone to bias. Most automated analyses of neurons rely on reconstruction of the neuron as a whole without a semantic analysis of each neurite. A fully-automated classification of all neurites still remains unavailable in open-source software. Here we present a standalone, GUI-based software for batch-quantification of neuronal morphology in two-dimensional fluorescence micrographs of cultured neurons with minimal requirements for user interaction. Single neurons are first reconstructed into binarized images using a Hessian-based segmentation algorithm to detect thin neurite structures combined with intensity- and shape-based reconstruction of the cell body. Neurites are then classified into axon, dendrites and their branches of increasing order using a geodesic distance transform of the cell skeleton. The software was benchmarked against a published dataset and reproduced the phenotype observed after manual annotation. Our tool promises accelerated and improved morphometric studies of neuronal morphology by allowing for consistent and automated analysis of large datasets.
Collapse
Affiliation(s)
- Amin Zehtabian
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany.
| | - Joachim Fuchs
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Molecular Biology and Biochemistry, Virchowweg 6, 10117 Berlin, Germany
| | - Britta J Eickholt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Molecular Biology and Biochemistry, Virchowweg 6, 10117 Berlin, Germany
| | - Helge Ewers
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany
| |
Collapse
|
5
|
He T, Hu C, Li S, Fan Y, Xie F, Sun X, Jiang Q, Chen W, Jia Y, Li W. The role of CD8 + T-cells in colorectal cancer immunotherapy. Heliyon 2024; 10:e33144. [PMID: 39005910 PMCID: PMC11239598 DOI: 10.1016/j.heliyon.2024.e33144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Immunotherapy has been an advanced and effective approach to treating various types of solid tumors in recent years, and the most successful strategy is immune checkpoint inhibitors (ICIs), which have shown beneficial effects in patients with colorectal cancer (CRC). Drug resistance to ICIs is usually associated with CD8+ T-cells targeting tumor antigens; thus, CD8+ T-cells play an important role in immunotherapy. Unfortunately, Under continuous antigen stimulation, tumor microenvironment(TME), hypoxia and other problems it leads to insufficient infiltration of CD8+ T-cells, low efficacy and mechanism exhaustion, which have become obstacles to immunotherapy. Thus, this article describes the relationship between CRC and the immune system, focuses on the process of CD8+ T-cells production, activation, transport, killing, and exhaustion, and expounds on related mechanisms leading to CD8+ T-cells exhaustion. Finally, this article summarizes the latest strategies and methods in recent years, focusing on improving the infiltration, efficacy, and exhaustion of CD8+ T-cells, which may help to overcome the barriers to immunotherapy.
Collapse
Affiliation(s)
- Tao He
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Chencheng Hu
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Shichao Li
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yao Fan
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Fei Xie
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Xin Sun
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Qingfeng Jiang
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Weidong Chen
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yingtian Jia
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Wusheng Li
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| |
Collapse
|
6
|
Volkov VA, Akhmanova A. Phase separation on microtubules: from droplet formation to cellular function? Trends Cell Biol 2024; 34:18-30. [PMID: 37453878 DOI: 10.1016/j.tcb.2023.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
Microtubules are cytoskeletal polymers that play important roles in numerous cellular processes, ranging from the control of cell shape and polarity to cell division and intracellular transport. Many of these roles rely on proteins that bind to microtubule ends and shafts, carry intrinsically disordered regions, and form complex multivalent interaction networks. A flurry of recent studies demonstrated that these properties allow diverse microtubule-binding proteins to undergo liquid-liquid phase separation (LLPS) in vitro. It is proposed that LLPS could potentially affect multiple microtubule-related processes, such as microtubule nucleation, control of microtubule dynamics and organization, and microtubule-based transport. Here, we discuss the evidence in favor and against the occurrence of LLPS and its functional significance for microtubule-based processes in cells.
Collapse
Affiliation(s)
- Vladimir A Volkov
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK.
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, Utrecht 3584 CH, The Netherlands.
| |
Collapse
|
7
|
Bensel BM, Previs S, Bookwalter C, Trybus KM, Walcott S, Warshaw DM. "Spatial Relationships Matter: Kinesin-1 Molecular Motors Transport Liposome Cargo Through 3D Microtubule Intersections In Vitro". BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569616. [PMID: 38076816 PMCID: PMC10705568 DOI: 10.1101/2023.12.01.569616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Kinesin-1 ensembles maneuver vesicular cargoes through intersections in the 3-dimensional (3D) intracellular microtubule (MT) network. To characterize directional outcomes (straight, turn, terminate) at MT intersections, we challenge 350 nm fluid-like liposomes transported by ~10 constitutively active, truncated kinesin-1 KIF5B (K543) with perpendicular 2-dimensional (2D) and 3D intersections in vitro. Liposomes frequently pause at 2D and 3D intersections (~2s), suggesting that motor teams can simultaneously engage each MT and undergo a tug-of-war. Once resolved, the directional outcomes at 2D MT intersections have a straight to turn ratio of 1.1; whereas at 3D MT intersections, liposomes more frequently go straight (straight to turn ratio of 1.8), highlighting that spatial relationships at intersections bias directional outcomes. Using 3D super-resolution microscopy (STORM), we define the gap between intersecting MTs and the liposome azimuthal approach angle heading into the intersection. We develop an in silico model in which kinesin-1 motors diffuse on the liposome surface, simultaneously engage the intersecting MTs, generate forces and detach from MTs governed by the motors' mechanochemical cycle, and undergo a tug-of-war with the winning team determining the directional outcome in 3D. The model predicts that 1-3 motors typically engage the MT, consistent with optical trapping measurements. Modeled liposomes also predominantly go straight through 3D intersections over a range of intersection gaps and liposome approach angles, even when obstructed by the crossing MT. Our observations and modeling offer mechanistic insights into how cells might tune the MT cytoskeleton, cargo, and motors to modulate cargo transport.
Collapse
Affiliation(s)
- Brandon M Bensel
- Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT 05405
| | - Samantha Previs
- Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT 05405
| | - Carol Bookwalter
- Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT 05405
| | - Kathleen M Trybus
- Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT 05405
| | - Sam Walcott
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, MA 01609
| | - David M Warshaw
- Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT 05405
| |
Collapse
|
8
|
Gu X, Jia C, Wang J. Advances in Understanding the Molecular Mechanisms of Neuronal Polarity. Mol Neurobiol 2023; 60:2851-2870. [PMID: 36738353 DOI: 10.1007/s12035-023-03242-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
The establishment and maintenance of neuronal polarity are important for neural development and function. Abnormal neuronal polarity establishment commonly leads to a variety of neurodevelopmental disorders. Over the past three decades, with the continuous development and improvement of biological research methods and techniques, we have made tremendous progress in the understanding of the molecular mechanisms of neuronal polarity establishment. The activity of positive and negative feedback signals and actin waves are both essential in this process. They drive the directional transport and aggregation of key molecules of neuronal polarity, promote the spatiotemporal regulation of ordered and coordinated interactions of actin filaments and microtubules, stimulate the specialization and growth of axons, and inhibit the formation of multiple axons. In this review, we focus on recent advances in these areas, in particular the important findings about neuronal polarity in two classical models, in vitro primary hippocampal/cortical neurons and in vivo cortical pyramidal neurons, and discuss our current understanding of neuronal polarity..
Collapse
Affiliation(s)
- Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
9
|
Iwanski MK, Kapitein LC. Cellular cartography: Towards an atlas of the neuronal microtubule cytoskeleton. Front Cell Dev Biol 2023; 11:1052245. [PMID: 37035244 PMCID: PMC10073685 DOI: 10.3389/fcell.2023.1052245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Microtubules, one of the major components of the cytoskeleton, play a crucial role during many aspects of neuronal development and function, such as neuronal polarization and axon outgrowth. Consequently, the microtubule cytoskeleton has been implicated in many neurodevelopmental and neurodegenerative disorders. The polar nature of microtubules is quintessential for their function, allowing them to serve as tracks for long-distance, directed intracellular transport by kinesin and dynein motors. Most of these motors move exclusively towards either the plus- or minus-end of a microtubule and some have been shown to have a preference for either dynamic or stable microtubules, those bearing a particular post-translational modification or those decorated by a specific microtubule-associated protein. Thus, it becomes important to consider the interplay of these features and their combinatorial effects on transport, as well as how different types of microtubules are organized in the cell. Here, we discuss microtubule subsets in terms of tubulin isotypes, tubulin post-translational modifications, microtubule-associated proteins, microtubule stability or dynamicity, and microtubule orientation. We highlight techniques used to study these features of the microtubule cytoskeleton and, using the information from these studies, try to define the composition, role, and organization of some of these subsets in neurons.
Collapse
Affiliation(s)
| | - Lukas C. Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
10
|
Wu Q, Yue X, Liu H, Zhu Y, Ke H, Yang X, Yin S, Li Z, Zhang Y, Hu T, Lan P, Wu X. MAP7D2 reduces CD8 + cytotoxic T lymphocyte infiltration through MYH9-HMGB1 axis in colorectal cancer. Mol Ther 2023; 31:90-104. [PMID: 36081350 PMCID: PMC9840115 DOI: 10.1016/j.ymthe.2022.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/17/2022] [Accepted: 09/06/2022] [Indexed: 01/28/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) represent a new paradigm in cancer immunotherapy, but can be largely restricted by the limited presence of CD8+ cytotoxic T lymphocytes (CTLs) in colorectal cancer (CRC) patients with microsatellite stable (MSS) tumors. Here, through next-generation sequencing, we identify microtubule-associated protein 7 domain 2 (MAP7D2) as an exploitable therapeutic maneuver to improve the efficacy of ICIs for MSS CRC therapy. In human CRC tissues, MAP7D2 expression is significantly increased in MSS CRC, and MAP7D2 adversely correlates with the presence of antitumor T lymphocytes. In vitro and in vivo experiments demonstrate that MAP7D2 knockdown significantly increases the infiltration of CD8+ CTLs, thereby inhibiting tumor progression and improving the efficacy of ICIs in MSS CRC murine models. Mechanistically, MAP7D2 interacts with MYH9 and protects it from ubiquitin-mediated degradation, subsequently decreasing the secretion of HMGB1, which suppresses the infiltration of CD8+ CTLs in MSS CRC. These findings highlight the importance of MAP7D2 in determining the infiltration of CD8+ CTLs and indicate that targeting MAP7D2 in MSS CRC may present a novel antitumor immunotherapy.
Collapse
Affiliation(s)
- Qian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao Yue
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huashan Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yaxi Zhu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Haoxian Ke
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Yang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shi Yin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhihao Li
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yunfeng Zhang
- Department of the General Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Tuo Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Xianrui Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor, Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Zhao Y, Huang S, Tan X, Long L, He Q, Liang X, Bai J, Li Q, Lin J, Li Y, Liu N, Ma J, Chen Y. N 6 -Methyladenosine-Modified CBX1 Regulates Nasopharyngeal Carcinoma Progression Through Heterochromatin Formation and STAT1 Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2205091. [PMID: 36310139 PMCID: PMC9798977 DOI: 10.1002/advs.202205091] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Indexed: 05/16/2023]
Abstract
Epitranscriptomic remodeling such as N6 -methyladenosine (m6 A) modification plays a critical role in tumor development. However, little is known about the underlying mechanisms connecting m6 A modification and nasopharyngeal carcinoma (NPC) progression. Here, CBX1 is identified, a histone methylation regulator, to be significantly upregulated with m6 A hypomethylation in metastatic NPC tissues. The m6 A-modified CBX1 mRNA transcript is recognized and destabilized by the m6 A reader YTHDF3. Furthermore, it is revealed that CBX1 promotes NPC cell migration, invasion, and proliferation through transcriptional repression of MAP7 via H3K9me3-mediated heterochromatin formation. In addition to its oncogenic effect, CBX1 can facilitate immune evasion through IFN-γ-STAT1 signaling-mediated PD-L1 upregulation. Clinically, CBX1 serves as an independent predictor for unfavorable prognosis in NPC patients. The results reveal a crosstalk between epitranscriptomic and epigenetic regulation in NPC progression, and shed light on the functions of CBX1 in tumorigenesis and immunomodulation, which may provide an appealing therapeutic target in NPC.
Collapse
Affiliation(s)
- Yin Zhao
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Shengyan Huang
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Xirong Tan
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Liufen Long
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Qingmei He
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Xiaoyu Liang
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Jiewen Bai
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Qingjie Li
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Jiayi Lin
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Yingqin Li
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Na Liu
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Jun Ma
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| | - Yupei Chen
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy651 Dongfeng Road EastGuangzhouGuangdong510060China
| |
Collapse
|
12
|
Chernov AV, Shubayev VI. Sexually dimorphic transcriptional programs of early-phase response in regenerating peripheral nerves. Front Mol Neurosci 2022; 15:958568. [PMID: 35983069 PMCID: PMC9378824 DOI: 10.3389/fnmol.2022.958568] [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: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
The convergence of transcriptional and epigenetic changes in the peripheral nervous system (PNS) reshapes the spatiotemporal gene expression landscape in response to nerve transection. The control of these molecular programs exhibits sexually dimorphic characteristics that remain not sufficiently characterized. In the present study, we recorded genome-wide and sex-dependent early-phase transcriptional changes in regenerating (proximal) sciatic nerve 24 h after axotomy. Male nerves exhibited more extensive transcriptional changes with male-dominant upregulation of cytoskeletal binding and structural protein genes. Regulation of mRNAs encoding ion and ionotropic neurotransmitter channels displayed prominent sexual dimorphism consistent with sex-specific mRNA axonal transport in an early-phase regenerative response. Protein kinases and axonal transport genes showed sexually dimorphic regulation. Genes encoding components of synaptic vesicles were at high baseline expression in females and showed post-injury induction selectively in males. Predictive bioinformatic analyses established patterns of sexually dimorphic regulation of neurotrophic and immune genes, including activation of glial cell line-derived neurotrophic factor Gfra1 receptor and immune checkpoint cyclin D1 (Ccnd1) potentially linked to X-chromosome encoded tissue inhibitor of matrix metallo proteinases 1 (Timp1). Regulatory networks involving Olig1, Pou3f3/Oct6, Myrf, and Myt1l transcription factors were linked to sex-dependent reprogramming in regenerating nerves. Differential expression patterns of non-coding RNAs motivate a model of sexually dimorphic nerve regenerative responses to injury determined by epigenetic factors. Combined with our findings in the corresponding dorsal root ganglia (DRG), unique early-phase sex-specific molecular triggers could enrich the mechanistic understanding of peripheral neuropathies.
Collapse
Affiliation(s)
- Andrei V. Chernov
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, United States
- VA San Diego Healthcare System, San Diego, CA, United States
- *Correspondence: Andrei V. Chernov,
| | - Veronica I. Shubayev
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, United States
- VA San Diego Healthcare System, San Diego, CA, United States
| |
Collapse
|
13
|
Lin G, Lin H, Zhuo R, He W, Ma C, Liu Y, Liu M. GCN5/KAT2A contributes to axon growth and neurogenesis. Neurosci Lett 2022; 784:136742. [PMID: 35716963 DOI: 10.1016/j.neulet.2022.136742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/31/2022] [Accepted: 06/13/2022] [Indexed: 10/18/2022]
Abstract
Posttranslational modification (PTM) of tubulin proteins is involved in microtubule dynamics. Acetylation, an important alpha-tubulin PTM, which is regarded as a hallmark event of stable microtubules, often occurs in neurogenesis and axon outgrowth. GCN5/KAT2A is a well-known histone acetyltransferase and has also been reported to hold the activity of nonhistone acetyltransferases, such as acetylated tubulin (Ace-tubulin). In this study, we investigated the role of GCN5/KAT2A in axon growth and neurogenesis. E18 cortical neurons obtained from day 18 embryos of pregnant Sprague-Dawley (SD) rats were cultured and transfected with GCN5 siRNA or treated with the GCN5 inhibitor MB-3. Neural stem cells (NSCs) derived from the cerebral cortexes of E14 SD rats were cultured and differentiated. During differentiation, MB-3 was applied to investigate the effect of GCN5 dysfunction on neurogenesis. The axonal length and the ratio and distribution of acetylated and tyrosinated tubulin (Tyr-tubulin) were evaluated by immunostaining assay. The expression levels of Nestin, Tuj1, acetylated tubulin, and tyrosinated tubulin proteins were analyzed by Western blotting assays. In primary neurons, both GCN5 siRNA and MB-3 treatment reduced acetylated tubulin protein, changed the ratio of acetylated and tyrosinated tubulin, and decreased axonal length. During NSC differentiation, MB-3 application reduced axon outgrowth, decreased acetylated tubulin and altered the distribution of acetylated tubulin and tyrosinated tubulin. This study revealed for the first time that the acetyltransferase GCN5/KAT2A could contribute to axon outgrowth by altering the ratio and distribution of acetylated tubulin.
Collapse
Affiliation(s)
- Ge Lin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Haixu Lin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Run Zhuo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Wei He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Chao Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China.
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China.
| |
Collapse
|
14
|
Population-scale analysis of common and rare genetic variation associated with hearing loss in adults. Commun Biol 2022; 5:540. [PMID: 35661827 PMCID: PMC9166757 DOI: 10.1038/s42003-022-03408-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/23/2022] [Indexed: 12/29/2022] Open
Abstract
To better understand the genetics of hearing loss, we performed a genome-wide association meta-analysis with 125,749 cases and 469,497 controls across five cohorts. We identified 53/c loci affecting hearing loss risk, including common coding variants in COL9A3 and TMPRSS3. Through exome sequencing of 108,415 cases and 329,581 controls, we observed rare coding associations with 11 Mendelian hearing loss genes, including additive effects in known hearing loss genes GJB2 (Gly12fs; odds ratio [OR] = 1.21, P = 4.2 × 10-11) and SLC26A5 (gene burden; OR = 1.96, P = 2.8 × 10-17). We also identified hearing loss associations with rare coding variants in FSCN2 (OR = 1.14, P = 1.9 × 10-15) and KLHDC7B (OR = 2.14, P = 5.2 × 10-30). Our results suggest a shared etiology between Mendelian and common hearing loss in adults. This work illustrates the potential of large-scale exome sequencing to elucidate the genetic architecture of common disorders where both common and rare variation contribute to risk.
Collapse
|
15
|
Chiba K, Ori-McKenney KM, Niwa S, McKenney RJ. Synergistic autoinhibition and activation mechanisms control kinesin-1 motor activity. Cell Rep 2022; 39:110900. [PMID: 35649356 PMCID: PMC9365671 DOI: 10.1016/j.celrep.2022.110900] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/22/2022] [Accepted: 05/10/2022] [Indexed: 11/03/2022] Open
Abstract
Kinesin-1 activity is regulated by autoinhibition. Intramolecular interactions within the kinesin heavy chain (KHC) are proposed to be one facet of motor regulation. The KHC also binds to the kinesin light chain (KLC), which has been implicated in both autoinhibition and activation of the motor. We show that the KLC inhibits the kinesin-microtubule interaction independently from the proposed intramolecular interaction within KHC. Cargo-adaptor proteins that bind the KLC stimulated processive movement, but the landing rate of activated kinesin complexes remained low. Mitogen-activated protein 7 (MAP7) enhanced motility by increasing the landing rate and run length of the activated kinesin motors. Our results support a model whereby the motor activity of the kinesin is regulated by synergistic inhibition mechanisms and that cargo-adaptor binding to the KLC releases both mechanisms. However, a non-motor MAP is required for robust microtubule association of the activated motor. Thus, human kinesin is regulated by synergistic autoinhibition and activation mechanisms.
Collapse
Affiliation(s)
- Kyoko Chiba
- Department of Molecular and Cellular Biology, University of California, Davis, 145 Briggs Hall, Davis, CA 95616, USA; Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Miyagi, 6-3 Aramaki Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Kassandra M Ori-McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, 145 Briggs Hall, Davis, CA 95616, USA
| | - Shinsuke Niwa
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Miyagi, 6-3 Aramaki Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Richard J McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, 145 Briggs Hall, Davis, CA 95616, USA.
| |
Collapse
|
16
|
Kikuchi K, Sakamoto Y, Uezu A, Yamamoto H, Ishiguro KI, Shimamura K, Saito T, Hisanaga SI, Nakanishi H. Map7D2 and Map7D1 facilitate microtubule stabilization through distinct mechanisms in neuronal cells. Life Sci Alliance 2022; 5:5/8/e202201390. [PMID: 35470240 PMCID: PMC9039348 DOI: 10.26508/lsa.202201390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 12/05/2022] Open
Abstract
The microtubule-associated proteins Map7D2 and Map7D1, which belong to the MAP7 family, stabilize microtubules through distinct mechanisms for the control of cell motility and neurite outgrowth. Microtubule (MT) dynamics are modulated through the coordinated action of various MT-associated proteins (MAPs). However, the regulatory mechanisms underlying MT dynamics remain unclear. We show that the MAP7 family protein Map7D2 stabilizes MTs to control cell motility and neurite outgrowth. Map7D2 directly bound to MTs through its N-terminal half and stabilized MTs in vitro. Map7D2 localized prominently to the centrosome and partially on MTs in mouse N1-E115 neuronal cells, which expresses two of the four MAP7 family members, Map7D2 and Map7D1. Map7D2 loss decreased the resistance to the MT-destabilizing agent nocodazole without affecting acetylated/detyrosinated stable MTs, suggesting that Map7D2 stabilizes MTs via direct binding. In addition, Map7D2 loss increased the rate of random cell migration and neurite outgrowth, presumably by disturbing the balance between MT stabilization and destabilization. Map7D1 exhibited similar subcellular localization and gene knockdown phenotypes to Map7D2. However, in contrast to Map7D2, Map7D1 was required for the maintenance of acetylated stable MTs. Taken together, our data suggest that Map7D2 and Map7D1 facilitate MT stabilization through distinct mechanisms in cell motility and neurite outgrowth.
Collapse
Affiliation(s)
- Koji Kikuchi
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhisa Sakamoto
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akiyoshi Uezu
- Department of Cell Biology, Duke University Medical School, Durham, NC, USA
| | - Hideyuki Yamamoto
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kei-Ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Kenji Shimamura
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Taro Saito
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Japan
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Japan
| | - Hiroyuki Nakanishi
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
17
|
Tymanskyj SR, Curran BM, Ma L. Selective axonal transport through branch junctions is directed by growth cone signaling and mediated by KIF1/kinesin-3 motors. Cell Rep 2022; 39:110748. [PMID: 35476993 PMCID: PMC9097860 DOI: 10.1016/j.celrep.2022.110748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/27/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Development and function of nerve cells rely on the orchestration of microtubule-based transport from the cell body into distal axonal terminals. Neurons often have highly elaborate branches innervating multiple targets, but how protein or membrane cargos navigate through branch junctions to specific branch targets is unknown. Here, we demonstrate that anterograde transport of membrane vesicles through axonal branch junctions is highly selective, which is influenced by branch length and more strongly by growth cone motility. Using an optogenetic tool, we demonstrate that signaling from the growth cone can rapidly direct transport through branch junctions. We further demonstrate that such transport selectivity is differentially regulated for different vesicles and mediated by the KIF1/kinesin-3 family motors. We propose that this transport regulation through branch junctions could broadly impact neuronal development, function, and regeneration.
Collapse
Affiliation(s)
- Stephen R Tymanskyj
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bridget M Curran
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Le Ma
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
18
|
Fuchs J, Eickholt BJ. Precursor types predict the stability of neuronal branches. J Cell Sci 2021; 134:273430. [PMID: 34766183 PMCID: PMC8714070 DOI: 10.1242/jcs.258983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/03/2021] [Indexed: 11/20/2022] Open
Abstract
Branches are critical for neuron function, generating the morphological complexity required for functional networks. They emerge from different, well-described, cytoskeletal precursor structures that elongate to branches. While branches are thought to be maintained by shared cytoskeletal regulators, our data from mouse hippocampal neurons indicate that the precursor structures trigger alternative branch maintenance mechanisms with differing stabilities. Whereas branches originating from lamellipodia or growth cone splitting events collapse soon after formation, branches emerging from filopodia persist. Furthermore, compared to other developing neurites, axons stabilise all branches and preferentially initiate branches from filopodia. These differences explain the altered stability of branches we observe in neurons lacking the plasma membrane protein phospholipid phosphatase-related protein 3 (PLPPR3, also known as PRG2) and in neurons treated with netrin-1. Rather than altering branch stability directly, PLPPR3 and netrin-1 boost a 'filopodia branch programme' on axons, thereby indirectly initiating more long-lived branches. In summary, we propose that studies on branching should distinguish overall stabilising effects from effects on precursor types, ideally using multifactorial statistical models, as exemplified in this study.
Collapse
Affiliation(s)
- Joachim Fuchs
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Molecular Biology and Biochemistry, Virchowweg 6, 10117 Berlin, Germany
| | - Britta J Eickholt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Molecular Biology and Biochemistry, Virchowweg 6, 10117 Berlin, Germany
| |
Collapse
|
19
|
Zhang W, Ciorraga M, Mendez P, Retana D, Boumedine-Guignon N, Achón B, Russier M, Debanne D, Garrido JJ. Formin Activity and mDia1 Contribute to Maintain Axon Initial Segment Composition and Structure. Mol Neurobiol 2021; 58:6153-6169. [PMID: 34458961 PMCID: PMC8639558 DOI: 10.1007/s12035-021-02531-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/11/2021] [Indexed: 10/29/2022]
Abstract
The axon initial segment (AIS) is essential for maintaining neuronal polarity, modulating protein transport into the axon, and action potential generation. These functions are supported by a distinctive actin and microtubule cytoskeleton that controls axonal trafficking and maintains a high density of voltage-gated ion channels linked by scaffold proteins to the AIS cytoskeleton. However, our knowledge of the mechanisms and proteins involved in AIS cytoskeleton regulation to maintain or modulate AIS structure is limited. In this context, formins play a significant role in the modulation of actin and microtubules. We show that pharmacological inhibition of formins modifies AIS actin and microtubule characteristics in cultured hippocampal neurons, reducing F-actin density and decreasing microtubule acetylation. Moreover, formin inhibition diminishes sodium channels, ankyrinG and βIV-spectrin AIS density, and AIS length, in cultured neurons and brain slices, accompanied by decreased neuronal excitability. We show that genetic downregulation of the mDia1 formin by interference RNAs also decreases AIS protein density and shortens AIS length. The ankyrinG decrease and AIS shortening observed in pharmacologically inhibited neurons and neuron-expressing mDia1 shRNAs were impaired by HDAC6 downregulation or EB1-GFP expression, known to increase microtubule acetylation or stability. However, actin stabilization only partially prevented AIS shortening without affecting AIS protein density loss. These results suggest that mDia1 maintain AIS composition and length contributing to the stability of AIS microtubules.
Collapse
Affiliation(s)
- Wei Zhang
- Instituto Cajal, CSIC, 28002 Madrid, Spain
- Present Address: College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | | | | | | | | | | | - Michaël Russier
- UNIS, INSERM, UMR 1072, Aix-Marseille Université, 13015 Marseille, France
| | - Dominique Debanne
- UNIS, INSERM, UMR 1072, Aix-Marseille Université, 13015 Marseille, France
| | - Juan José Garrido
- Instituto Cajal, CSIC, 28002 Madrid, Spain
- Alzheimer’s Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
20
|
Network Analysis Identifies Sex-Specific Gene Expression Changes in Blood of Amyotrophic Lateral Sclerosis Patients. Int J Mol Sci 2021; 22:ijms22137150. [PMID: 34281203 PMCID: PMC8269377 DOI: 10.3390/ijms22137150] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 01/18/2023] Open
Abstract
Understanding the molecular mechanisms underlying the pathogenesis of amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease, is a major challenge. We used co-expression networks implemented by the SWitch Miner software to identify switch genes associated with drastic transcriptomic changes in the blood of ALS patients. Functional analyses revealed that switch genes were enriched in pathways related to the cell cycle, hepatitis C, and small cell lung cancer. Analysis of switch genes by sex revealed that switch genes from males were associated with metabolic pathways, including PI3K-AKT, sphingolipid, carbon metabolism, FOXO, and AMPK signaling. In contrast, female switch genes related to infectious diseases, inflammation, apoptosis, and atherosclerosis. Furthermore, eight switch genes showed sex-specific gene expression patterns. Collectively, we identified essential genes and pathways that may explain sex differences observed in ALS. Future studies investigating the potential role of these genes in driving disease disparities between males and females with ALS are warranted.
Collapse
|
21
|
Multiple layers of spatial regulation coordinate axonal cargo transport. Curr Opin Neurobiol 2021; 69:241-246. [PMID: 34171618 DOI: 10.1016/j.conb.2021.03.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 11/23/2022]
Abstract
Nerve axons are shaped similar to long electric wires to quickly transmit information from one end of the body to the other. To remain healthy and functional, axons depend on a wide range of cellular cargos to be transported from the neuronal cell body to its distal processes. Because of the extended distance, a sophisticated and well-organized trafficking network is required to move cargos up and down the axon. Besides motor proteins driving cargo transport, recent data revealed that subcellular membrane specializations, including the axon initial segment at the beginning of the axon and the membrane-associated periodic skeleton, which extends throughout the axonal length, are important spatial regulators of cargo traffic. In addition, tubulin modifications and microtubule-associated proteins present along the axonal cytoskeleton have been proposed to bias cargo movements. Here, we discuss the recent advances in understanding these multiple layers of regulatory mechanisms controlling axonal transport.
Collapse
|
22
|
Wang J, Lan Y, He L, Tang R, Li Y, Huang Y, Liang S, Gao Z, Price M, Yue B, He M, Guo T, Fan Z. Sex-specific gene expression in the blood of four primates. Genomics 2021; 113:2605-2613. [PMID: 34116169 DOI: 10.1016/j.ygeno.2021.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/03/2021] [Accepted: 06/06/2021] [Indexed: 10/21/2022]
Abstract
Blood is an important non-reproductive tissue, but little is known about the sex-specific gene expressions in the blood. Therefore, we investigated sex-specific gene expression differences in the blood tissues of four primates, rhesus macaques (Macaca mulatta), Tibetan macaques (M. thibetana), yellow baboons (Papio cynocephalus), and humans. We identified seven sex-specific differentially expressed genes (SDEGs) in each non-human primate and 31 SDEGs in humans. The four primates had only one common SDEG, MAP7D2. In humans, immune-related SDEGs were identified as up-regulated, but also down-regulated in females. We also found that most of the X-Y gene pairs had similar expression levels between species, except pair EIF1AY/EIF1AX. The expression level of X-Y gene pairs of rhesus and Tibetan macaques showed no significant differential expression levels, while humans had six significant XY-biased and three XX-biased X-Y gene pairs. Our observed sex differences in blood should increase understanding of sex differences in primate blood tissue.
Collapse
Affiliation(s)
- Jiao Wang
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China
| | - Yue Lan
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China
| | - Lewei He
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China
| | - Ruixiang Tang
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610064, Sichuan, China
| | - Yuhui Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu 610052, Sichuan, China
| | - Yuan Huang
- Medical Laboratory Department of West China Fourth Hospital, Sichuan University, Chengdu 610000, Sichuan, China
| | - Shan Liang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu 610052, Sichuan, China
| | - Zhan Gao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu 610052, Sichuan, China
| | - Megan Price
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China.
| | - Bisong Yue
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Miao He
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu 610052, Sichuan, China.
| | - Tao Guo
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China.
| |
Collapse
|
23
|
Fu X, An Y, Wang H, Li P, Lin J, Yuan J, Yue R, Jin Y, Gao J, Chai R. Deficiency of Klc2 Induces Low-Frequency Sensorineural Hearing Loss in C57BL/6 J Mice and Human. Mol Neurobiol 2021; 58:4376-4391. [PMID: 34014435 DOI: 10.1007/s12035-021-02422-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022]
Abstract
The transport system in cochlear hair cells (HCs) is important for their function, and the kinesin family of proteins transports numerous cellular cargos via the microtubule network in the cytoplasm. Here, we found that Klc2 (kinesin light chain 2), the light chain of kinesin-1 that mediates cargo binding and regulates kinesin-1 motility, is essential for cochlear function. We generated mice lacking Klc2, and they suffered from low-frequency hearing loss as early as 1 month of age. We demonstrated that deficiency of Klc2 resulted in abnormal transport of mitochondria and the down-regulation of the GABAA receptor family. In addition, whole-genome sequencing (WGS) of patient showed that KLC2 was related to low-frequency hearing in human. Hence, to explore therapeutic approaches, we developed adeno-associated virus containing the Klc2 wide-type cDNA sequence, and Klc2-null mice delivered virus showed apparent recovery, including decreased ABR threshold and reduced out hair cell (OHC) loss. In summary, we show that the kinesin transport system plays an indispensable and special role in cochlear HC function in mice and human and that mitochondrial localization is essential for HC survival.
Collapse
Affiliation(s)
- Xiaolong Fu
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,College of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yachun An
- School of Life Science, Shandong University, Qingdao, China
| | - Hongyang Wang
- College of Otolaryngology, Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China
| | - Peipei Li
- School of Life Science, Shandong University, Qingdao, China
| | - Jing Lin
- Waksman Institute, the State University of New Jersey, RutgersNew Brunswick, NJ, USA
| | - Jia Yuan
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Rongyu Yue
- Department of Otolaryngology-Head and Neck Surgery, Provincial Hospital Affiliated To Shandong University, Jinan, China
| | - Yecheng Jin
- School of Life Science, Shandong University, Qingdao, China
| | - Jiangang Gao
- College of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China. .,College of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
| |
Collapse
|
24
|
Shah M, Chacko LA, Joseph JP, Ananthanarayanan V. Mitochondrial dynamics, positioning and function mediated by cytoskeletal interactions. Cell Mol Life Sci 2021; 78:3969-3986. [PMID: 33576841 PMCID: PMC11071877 DOI: 10.1007/s00018-021-03762-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/27/2020] [Accepted: 01/15/2021] [Indexed: 12/22/2022]
Abstract
The ability of a mitochondrion to undergo fission and fusion, and to be transported and localized within a cell are central not just to proper functioning of mitochondria, but also to that of the cell. The cytoskeletal filaments, namely microtubules, F-actin and intermediate filaments, have emerged as prime movers in these dynamic mitochondrial shape and position transitions. In this review, we explore the complex relationship between the cytoskeleton and the mitochondrion, by delving into: (i) how the cytoskeleton helps shape mitochondria via fission and fusion events, (ii) how the cytoskeleton facilitates the translocation and anchoring of mitochondria with the activity of motor proteins, and (iii) how these changes in form and position of mitochondria translate into functioning of the cell.
Collapse
Affiliation(s)
- Mitali Shah
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Leeba Ann Chacko
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Joel P Joseph
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Vaishnavi Ananthanarayanan
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India.
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
25
|
Koppers M, Farías GG. Organelle distribution in neurons: Logistics behind polarized transport. Curr Opin Cell Biol 2021; 71:46-54. [PMID: 33706233 DOI: 10.1016/j.ceb.2021.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 12/20/2022]
Abstract
Highly polarized neurons need to carefully regulate the distribution of organelles and other cargoes into their two morphologically and functionally distinct domains, the somatodendritic and axonal compartments, to maintain proper neuron homeostasis. An outstanding question in the field is how organelles reach their correct destination. Long-range transport along microtubules, driven by motors, ensures a fast and controlled availability of organelles in axons and dendrites, but it remains largely unclear what rules govern their transport into the correct compartment. Here, we review the emerging concepts of polarized cargo trafficking in neurons, highlighting the role of microtubule organization, microtubule-associated proteins, and motor proteins and discuss compartment-specific inclusion and exclusion mechanisms as well as the regulation of correct coupling of cargoes to motor proteins.
Collapse
Affiliation(s)
- Max Koppers
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
| | - Ginny G Farías
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands.
| |
Collapse
|
26
|
Zahavi EE, Hummel JJA, Han Y, Bar C, Stucchi R, Altelaar M, Hoogenraad CC. Combined kinesin-1 and kinesin-3 activity drives axonal trafficking of TrkB receptors in Rab6 carriers. Dev Cell 2021; 56:494-508.e7. [PMID: 33571451 PMCID: PMC7907685 DOI: 10.1016/j.devcel.2021.01.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 10/18/2020] [Accepted: 01/19/2021] [Indexed: 12/24/2022]
Abstract
Neurons depend on proper localization of neurotrophic receptors in their distal processes for their function. The Trk family of neurotrophin receptors controls neuronal survival, differentiation, and remodeling and are well known to function as retrograde signal carriers transported from the distal axon toward the cell body. However, the mechanism driving anterograde trafficking of Trk receptors into the axon is not well established. We used microfluidic compartmental devices and inducible secretion assay to systematically investigate the retrograde and anterograde trafficking routes of TrkB receptor along the axon in rat hippocampal neurons. We show that newly synthesized TrkB receptors traffic through the secretory pathway and are directly delivered into axon. We found that these TrkB carriers associate and are regulated by Rab6. Furthermore, the combined activity of kinesin-1 and kinesin-3 is needed for the formation of axon-bound TrkB secretory carriers and their effective entry and processive anterograde transport beyond the proximal axon.
Collapse
Affiliation(s)
- Eitan Erez Zahavi
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Jessica J A Hummel
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Yuhao Han
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Citlali Bar
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Riccardo Stucchi
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 Utrecht, the Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 Utrecht, the Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands; Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
27
|
Imbalanced post- and extrasynaptic SHANK2A functions during development affect social behavior in SHANK2-mediated neuropsychiatric disorders. Mol Psychiatry 2021; 26:6482-6504. [PMID: 34021263 PMCID: PMC8760046 DOI: 10.1038/s41380-021-01140-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 02/04/2023]
Abstract
Mutations in SHANK genes play an undisputed role in neuropsychiatric disorders. Until now, research has focused on the postsynaptic function of SHANKs, and prominent postsynaptic alterations in glutamatergic signal transmission have been reported in Shank KO mouse models. Recent studies have also suggested a possible presynaptic function of SHANK proteins, but these remain poorly defined. In this study, we examined how SHANK2 can mediate electrophysiological, molecular, and behavioral effects by conditionally overexpressing either wild-type SHANK2A or the extrasynaptic SHANK2A(R462X) variant. SHANK2A overexpression affected pre- and postsynaptic targets and revealed a reversible, development-dependent autism spectrum disorder-like behavior. SHANK2A also mediated redistribution of Ca2+-permeable AMPA receptors between apical and basal hippocampal CA1 dendrites, leading to impaired synaptic plasticity in the basal dendrites. Moreover, SHANK2A overexpression reduced social interaction and increased the excitatory noise in the olfactory cortex during odor processing. In contrast, overexpression of the extrasynaptic SHANK2A(R462X) variant did not impair hippocampal synaptic plasticity, but still altered the expression of presynaptic/axonal signaling proteins. We also observed an attention-deficit/hyperactivity-like behavior and improved social interaction along with enhanced signal-to-noise ratio in cortical odor processing. Our results suggest that the disruption of pre- and postsynaptic SHANK2 functions caused by SHANK2 mutations has a strong impact on social behavior. These findings indicate that pre- and postsynaptic SHANK2 actions cooperate for normal neuronal function, and that an imbalance between these functions may lead to different neuropsychiatric disorders.
Collapse
|
28
|
Serra-Marques A, Martin M, Katrukha EA, Grigoriev I, Peeters CAE, Liu Q, Hooikaas PJ, Yao Y, Solianova V, Smal I, Pedersen LB, Meijering E, Kapitein LC, Akhmanova A. Concerted action of kinesins KIF5B and KIF13B promotes efficient secretory vesicle transport to microtubule plus ends. eLife 2020; 9:e61302. [PMID: 33174839 PMCID: PMC7710357 DOI: 10.7554/elife.61302] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/10/2020] [Indexed: 01/30/2023] Open
Abstract
Intracellular transport relies on multiple kinesins, but it is poorly understood which kinesins are present on particular cargos, what their contributions are and whether they act simultaneously on the same cargo. Here, we show that Rab6-positive secretory vesicles are transported from the Golgi apparatus to the cell periphery by kinesin-1 KIF5B and kinesin-3 KIF13B, which determine the location of secretion events. KIF5B plays a dominant role, whereas KIF13B helps Rab6 vesicles to reach freshly polymerized microtubule ends, to which KIF5B binds poorly, likely because its cofactors, MAP7-family proteins, are slow in populating these ends. Sub-pixel localization demonstrated that during microtubule plus-end directed transport, both kinesins localize to the vesicle front and can be engaged on the same vesicle. When vesicles reverse direction, KIF13B relocates to the middle of the vesicle, while KIF5B shifts to the back, suggesting that KIF5B but not KIF13B undergoes a tug-of-war with a minus-end directed motor.
Collapse
Affiliation(s)
- Andrea Serra-Marques
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Maud Martin
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Eugene A Katrukha
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Ilya Grigoriev
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Cathelijn AE Peeters
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Qingyang Liu
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Peter Jan Hooikaas
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Yao Yao
- Departments of Medical Informatics and Radiology, Biomedical Imaging Group Rotterdam, Erasmus University Medical CenterRotterdamNetherlands
| | - Veronika Solianova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Ihor Smal
- Departments of Medical Informatics and Radiology, Biomedical Imaging Group Rotterdam, Erasmus University Medical CenterRotterdamNetherlands
| | - Lotte B Pedersen
- Department of Biology, Section of Cell Biology and Physiology, the August Krogh Building, University of CopenhagenCopenhagenDenmark
| | - Erik Meijering
- Departments of Medical Informatics and Radiology, Biomedical Imaging Group Rotterdam, Erasmus University Medical CenterRotterdamNetherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| |
Collapse
|
29
|
Vasudevan A, Koushika SP. Molecular mechanisms governing axonal transport: a C. elegans perspective. J Neurogenet 2020; 34:282-297. [PMID: 33030066 DOI: 10.1080/01677063.2020.1823385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Axonal transport is integral for maintaining neuronal form and function, and defects in axonal transport have been correlated with several neurological diseases, making it a subject of extensive research over the past several years. The anterograde and retrograde transport machineries are crucial for the delivery and distribution of several cytoskeletal elements, growth factors, organelles and other synaptic cargo. Molecular motors and the neuronal cytoskeleton function as effectors for multiple neuronal processes such as axon outgrowth and synapse formation. This review examines the molecular mechanisms governing axonal transport, specifically highlighting the contribution of studies conducted in C. elegans, which has proved to be a tractable model system in which to identify both novel and conserved regulatory mechanisms of axonal transport.
Collapse
Affiliation(s)
- Amruta Vasudevan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
30
|
Guillaud L, El-Agamy SE, Otsuki M, Terenzio M. Anterograde Axonal Transport in Neuronal Homeostasis and Disease. Front Mol Neurosci 2020; 13:556175. [PMID: 33071754 PMCID: PMC7531239 DOI: 10.3389/fnmol.2020.556175] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Neurons are highly polarized cells with an elongated axon that extends far away from the cell body. To maintain their homeostasis, neurons rely extensively on axonal transport of membranous organelles and other molecular complexes. Axonal transport allows for spatio-temporal activation and modulation of numerous molecular cascades, thus playing a central role in the establishment of neuronal polarity, axonal growth and stabilization, and synapses formation. Anterograde and retrograde axonal transport are supported by various molecular motors, such as kinesins and dynein, and a complex microtubule network. In this review article, we will primarily discuss the molecular mechanisms underlying anterograde axonal transport and its role in neuronal development and maturation, including the establishment of functional synaptic connections. We will then provide an overview of the molecular and cellular perturbations that affect axonal transport and are often associated with axonal degeneration. Lastly, we will relate our current understanding of the role of axonal trafficking concerning anterograde trafficking of mRNA and its involvement in the maintenance of the axonal compartment and disease.
Collapse
Affiliation(s)
- Laurent Guillaud
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Sara Emad El-Agamy
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Miki Otsuki
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
31
|
Radler MR, Suber A, Spiliotis ET. Spatial control of membrane traffic in neuronal dendrites. Mol Cell Neurosci 2020; 105:103492. [PMID: 32294508 PMCID: PMC7317674 DOI: 10.1016/j.mcn.2020.103492] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendrites are highly branched and specialized compartments with distinct structures and secretory organelles (e.g., spines, Golgi outposts), and a unique cytoskeletal organization that includes microtubules of mixed polarity. Dendritic membranes are enriched with proteins, which specialize in the formation and function of the post-synaptic membrane of the neuronal synapse. How these proteins partition preferentially in dendrites, and how they traffic in a manner that is spatiotemporally accurate and regulated by synaptic activity are long-standing questions of neuronal cell biology. Recent studies have shed new insights into the spatial control of dendritic membrane traffic, revealing new classes of proteins (e.g., septins) and cytoskeleton-based mechanisms with dendrite-specific functions. Here, we review these advances by revisiting the fundamental mechanisms that control membrane traffic at the levels of protein sorting and motor-driven transport on microtubules and actin filaments. Overall, dendrites possess unique mechanisms for the spatial control of membrane traffic, which might have specialized and co-evolved with their highly arborized morphology.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Ayana Suber
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Abstract
The intracellular transport system in neurons is specialized to an extraordinary degree, enabling the delivery of critical cargo to sites in axons or dendrites that are far removed from the cell center. Vesicles formed in the cell body are actively transported by kinesin motors along axonal microtubules to presynaptic sites that can be located more than a meter away. Both growth factors and degradative vesicles carrying aged organelles or aggregated proteins take the opposite route, driven by dynein motors. Distance is not the only challenge; precise delivery of cargos to sites of need must also be accomplished. For example, localized delivery of presynaptic components to hundreds of thousands of "en passant" synapses distributed along the length of a single axon in some neuronal subtypes provides a layer of complexity that must be successfully navigated to maintain synaptic transmission. We review recent advances in the field of axonal transport, with a focus on conceptual developments, and highlight our growing quantitative understanding of neuronal trafficking and its role in maintaining the synaptic function that underlies higher cognitive processes such as learning and memory.
Collapse
Affiliation(s)
- Pedro Guedes-Dias
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute of Neuronal Cell Biology, Technische Universität München, 80802 Munich, Germany
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Smart motors and cargo steering drive kinesin-mediated selective transport. Mol Cell Neurosci 2020; 103:103464. [PMID: 31972342 DOI: 10.1016/j.mcn.2019.103464] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Neurons are polarized cells, with dendrites and axons that require different complements of membrane proteins to fulfill their specialized functions. Membrane proteins are synthesized in the somatodendritic domain and delivered to their target membranes via long-range vesicle transport. Most anterograde vesicle transport is mediated by kinesin motors, but it is unclear how kinesins are targeted to axons or dendrites. Two main models have been proposed to explain kinesin selectivity. In the smart motor model, kinesin selectivity is conferred by a preference of the kinesin motor domain for specific subsets of microtubules. In the cargo steering model, kinesin selectivity is modulated by the vesicular cargo to which the motor is bound. We evaluate the evidence for both models and conclude that while the smart motor model may explain axonal selectivity, cargo steering is required for dendritic selectivity. Future work will determine the relative contributions of these models to polarized transport in living neurons.
Collapse
|
34
|
Luo Y, Xiang S, Hooikaas PJ, van Bezouwen L, Jijumon AS, Janke C, Förster F, Akhmanova A, Baldus M. Direct observation of dynamic protein interactions involving human microtubules using solid-state NMR spectroscopy. Nat Commun 2020; 11:18. [PMID: 31896752 PMCID: PMC6940360 DOI: 10.1038/s41467-019-13876-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
Microtubules are important components of the eukaryotic cytoskeleton. Their structural organization is regulated by nucleotide binding and many microtubule-associated proteins (MAPs). While cryo-EM and X-ray crystallography have provided detailed views of interactions between MAPs with the microtubule lattice, little is known about how MAPs and their intrinsically disordered regions interact with the dynamic microtubule surface. NMR carries the potential to directly probe such interactions but so far has been precluded by the low tubulin yield. We present a protocol to produce [13C, 15N]-labeled, functional microtubules (MTs) from human cells for solid-state NMR studies. This approach allowed us to demonstrate that MAPs can differently modulate the fast time-scale dynamics of C-terminal tubulin tails, suggesting distinct interaction modes. Our results pave the way for in-depth NMR studies of protein dynamics involved in MT assembly and their interactions with other cellular components.
Collapse
Affiliation(s)
- Yanzhang Luo
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - ShengQi Xiang
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
- MOE Key Lab for Membrane-less Organelles & Cellular Dynamics, School of Life Sciences, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, Anhui, China
| | - Peter Jan Hooikaas
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Laura van Bezouwen
- Cryo-Electron Microscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - A S Jijumon
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405, Orsay, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405, Orsay, France
| | - Friedrich Förster
- Cryo-Electron Microscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - Marc Baldus
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
35
|
MAP7 Prevents Axonal Branch Retraction by Creating a Stable Microtubule Boundary to Rescue Polymerization. J Neurosci 2019; 39:7118-7131. [PMID: 31391261 DOI: 10.1523/jneurosci.0775-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/16/2019] [Accepted: 07/31/2019] [Indexed: 01/07/2023] Open
Abstract
Complex neural circuits are built from axonal branches that allow each neuron to connect with multiple targets. During development, maturation of nascent branches depends on stabilization of newly assembled or transported microtubules, which are thought to be regulated by microtubule-associated proteins (MAPs). However, because many known MAPs inhibit branch formation, it is not clear which MAP is responsible for regulating microtubule stability during branch development. Here, we show that MAP7, a less-well understood MAP that is localized to branch junctions, provides a key molecular mechanism to regulate microtubule stability during branch formation. In developing rodent sensory neurons of mixed sex, MAP7 is required for branch maturation mainly by preventing branch retraction. This function is mediated by the ability of MAP7 to control microtubule stability, as microtubules are more stable at branch junctions where MAP7 is localized. Consistently, nascent branches depleted of MAP7 have decreased stable microtubules but increased dynamic microtubules. Moreover, MAP7 binds to the acetylated and stable region of individual microtubules and avoids the dynamic plus end, thereby creating a boundary that prevents microtubule depolymerization and rescues microtubule polymerization. This unique binding property, which is not observed for other MAPs, can prevent branch retraction caused by laser-induced severing or nocodazole-induced microtubule depolymerization. Together, our study identifies a novel molecular mechanism mediated by MAP7 to regulate microtubule stability and strengthen branches at different stages of axonal branch morphogenesis.SIGNIFICANCE STATEMENT Development and maintenance of axonal branches rely on microtubule stability, but the underlying molecular mechanisms are not fully understood. Here, we show that MAP7, a unique protein that interacts with both microtubules and the motor protein kinesin-1, plays a key role at branch junctions. MAP7 stabilizes microtubules in nascent branches and prevents branch retraction during branch maturation or after laser-induced injury. MAP7 also binds to the acetylated region of microtubules to prevent depolymerization and rescue polymerization. This unique binding property supports a novel mechanism mediated by MAP7 to cooperate with other MAPs and control microtubule stability during axonal branch development. This mechanism could also impact microtubule regulation in branch regeneration after nerve injury.
Collapse
|