51
|
MacTaggart B, Kashina A. Posttranslational modifications of the cytoskeleton. Cytoskeleton (Hoboken) 2021; 78:142-173. [PMID: 34152688 DOI: 10.1002/cm.21679] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
The cytoskeleton plays important roles in many essential processes at the cellular and organismal levels, including cell migration and motility, cell division, and the establishment and maintenance of cell and tissue architecture. In order to facilitate these varied functions, the main cytoskeletal components-microtubules, actin filaments, and intermediate filaments-must form highly diverse intracellular arrays in different subcellular areas and cell types. The question of how this diversity is conferred has been the focus of research for decades. One key mechanism is the addition of posttranslational modifications (PTMs) to the major cytoskeletal proteins. This posttranslational addition of various chemical groups dramatically increases the complexity of the cytoskeletal proteome and helps facilitate major global and local cytoskeletal functions. Cytoskeletal proteins undergo many PTMs, most of which are not well understood. Recent technological advances in proteomics and cell biology have allowed for the in-depth study of individual PTMs and their functions in the cytoskeleton. Here, we provide an overview of the major PTMs that occur on the main structural components of the three cytoskeletal systems-tubulin, actin, and intermediate filament proteins-and highlight the cellular function of these modifications.
Collapse
Affiliation(s)
- Brittany MacTaggart
- School of Veterinary Medicine, Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna Kashina
- School of Veterinary Medicine, Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
52
|
Melo-Hanchuk TD, Kobarg J. Polyglutamylase activity of tubulin tyrosine ligase-like 4 is negatively regulated by the never in mitosis gene A family kinase never in mitosis gene A -related kinase 5. World J Biol Chem 2021; 12:38-51. [PMID: 34084286 PMCID: PMC8160597 DOI: 10.4331/wjbc.v12.i3.38] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/06/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Tubulins, building blocks of microtubules, are modified substrates of diverse post-translational modifications including phosphorylation, polyglycylation and polyglutamylation. Polyglutamylation of microtubules, catalyzed by enzymes from the tubulin tyrosine ligase-like (TTLL) family, can regulate interactions with molecular motors and other proteins. Due to the diversity and functional importance of microtubule modifications, strict control of the TTLL enzymes has been suggested.
AIM To characterize the interaction between never in mitosis gene A-related kinase 5 (NEK5) and TTLL4 proteins and the effects of TTLL4 phosphorylation.
METHODS The interaction between NEK5 and TTLL4 was identified by yeast two-hybrid screening using the C-terminus of NEK5 (a.a. 260–708) as bait and confirmed by immunoprecipitation. The phosphorylation sites of TTLL4 were identified by mass spectrometry and point mutations were introduced.
RESULTS Here, we show that NEK5 interacts with TTLL4 and regulates its polyglutamylation activity. We further show that NEK5 can also interact with TTLL5 and TTLL7. The silencing of NEK5 increases the levels of polyglutamylation of proteins by increasing the activity of TTLL4. The same effects were observed after the expression of the catalytically inactive form of NEK5. This regulation of TTLL4 activity involves its phosphorylation at Y815 and S1136 amino acid residues.
CONCLUSION Our results demonstrate, for the first time, the regulation of TTLL activity through phosphorylation, pointing to NEK5 as a potential effector kinase. We also suggest a general control of tubulin polyglutamylation through NEK family members in human cells.
Collapse
Affiliation(s)
| | - Jörg Kobarg
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083-862, Brazil
| |
Collapse
|
53
|
Zhang K, Zhu X, Durst S, Hohenberger P, Han MJ, An G, Sahi VP, Riemann M, Nick P. A rice tubulin tyrosine ligase-like 12 protein affects the dynamic and orientation of microtubules. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2021; 63:848-864. [PMID: 33336892 DOI: 10.1111/jipb.13059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/14/2020] [Indexed: 06/12/2023]
Abstract
The detyrosination/retyrosination cycle is the most common post-translational modification of α-tubulin. Removal of the conserved C-terminal tyrosine of α-tubulin by a still elusive tubulin tyrosine carboxypeptidase, and religation of this tyrosine by a tubulin tyrosine ligase (TTL), are probably common to all eukaryotes. Interestingly, for plants, the only candidates qualifying as potential TTL homologs are the tubulin tyrosine ligase-like 12 proteins. To get insight into the biological functions of these potential TTL homologs, we cloned the rice TTL-like 12 protein (OsTTLL12) and generated overexpression OsTTLL12-RFP lines in both rice and tobacco BY-2 cells. We found, unexpectedly, that overexpression of this OsTTLL12-RFP increased the relative abundance of detyrosinated α-tubulin in both coleoptile and seminal root, correlated with more stable microtubules. This was independent of the respective orientation of cortical microtubule, and followed by correspondingly changing growth of coleoptiles and seminal roots. A perturbed organization of phragmoplast microtubules and disoriented cell walls were further characteristics of this phenotype. Thus, the elevated tubulin detyrosination in consequence of OsTTLL12 overexpression affects structural and dynamic features of microtubules, followed by changes in the axiality of cell plate deposition and, consequently, plant growth.
Collapse
Affiliation(s)
- Kunxi Zhang
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| | - Xin Zhu
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| | - Steffen Durst
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| | - Petra Hohenberger
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| | - Min-Jung Han
- Aptamer Initiative, Postech Biotech Center, Pohang University of Science and Technology, Pohang-si, Gyeongsangbuk-do, 37673, South Korea
| | - Gynheung An
- Department of Plant Molecular Systems Biotech, Kyung Hee University, Yongin, 446-701, South Korea
| | - Vaidurya P Sahi
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| | - Michael Riemann
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| | - Peter Nick
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| |
Collapse
|
54
|
Yu L, Liu P. Cytosolic DNA sensing by cGAS: regulation, function, and human diseases. Signal Transduct Target Ther 2021; 6:170. [PMID: 33927185 PMCID: PMC8085147 DOI: 10.1038/s41392-021-00554-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/17/2021] [Accepted: 03/08/2021] [Indexed: 12/16/2022] Open
Abstract
Sensing invasive cytosolic DNA is an integral component of innate immunity. cGAS was identified in 2013 as the major cytosolic DNA sensor that binds dsDNA to catalyze the synthesis of a special asymmetric cyclic-dinucleotide, 2'3'-cGAMP, as the secondary messenger to bind and activate STING for subsequent production of type I interferons and other immune-modulatory genes. Hyperactivation of cGAS signaling contributes to autoimmune diseases but serves as an adjuvant for anticancer immune therapy. On the other hand, inactivation of cGAS signaling causes deficiency to sense and clear the viral and bacterial infection and creates a tumor-prone immune microenvironment to facilitate tumor evasion of immune surveillance. Thus, cGAS activation is tightly controlled. In this review, we summarize up-to-date multilayers of regulatory mechanisms governing cGAS activation, including cGAS pre- and post-translational regulations, cGAS-binding proteins, and additional cGAS regulators such as ions and small molecules. We will also reveal the pathophysiological function of cGAS and its product cGAMP in human diseases. We hope to provide an up-to-date review for recent research advances of cGAS biology and cGAS-targeted therapies for human diseases.
Collapse
Affiliation(s)
- Le Yu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
55
|
Yang WT, Hong SR, He K, Ling K, Shaiv K, Hu J, Lin YC. The Emerging Roles of Axonemal Glutamylation in Regulation of Cilia Architecture and Functions. Front Cell Dev Biol 2021; 9:622302. [PMID: 33748109 PMCID: PMC7970040 DOI: 10.3389/fcell.2021.622302] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cilia, which either generate coordinated motion or sense environmental cues and transmit corresponding signals to the cell body, are highly conserved hair-like structures that protrude from the cell surface among diverse species. Disruption of ciliary functions leads to numerous human disorders, collectively referred to as ciliopathies. Cilia are mechanically supported by axonemes, which are composed of microtubule doublets. It has been recognized for several decades that tubulins in axonemes undergo glutamylation, a post-translational polymodification, that conjugates glutamic acid chains onto the C-terminal tail of tubulins. However, the physiological roles of axonemal glutamylation were not uncovered until recently. This review will focus on how cells modulate glutamylation on ciliary axonemes and how axonemal glutamylation regulates cilia architecture and functions, as well as its physiological importance in human health. We will also discuss the conventional and emerging new strategies used to manipulate glutamylation in cilia.
Collapse
Affiliation(s)
- Wen-Ting Yang
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - Shi-Rong Hong
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - Kai He
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Kritika Shaiv
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - JingHua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
- Department of Medical Science, National Tsing Hua University, HsinChu City, Taiwan
| |
Collapse
|
56
|
Bodakuntla S, Janke C, Magiera MM. Tubulin polyglutamylation, a regulator of microtubule functions, can cause neurodegeneration. Neurosci Lett 2021; 746:135656. [PMID: 33482309 DOI: 10.1016/j.neulet.2021.135656] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases lead to a progressive demise of neuronal functions that ultimately results in neuronal death. Besides a large variety of molecular pathways that have been linked to the degeneration of neurons, dysfunctions of the microtubule cytoskeleton are common features of many human neurodegenerative disorders. Yet, it is unclear whether microtubule dysfunctions are causative, or mere bystanders in the disease progression. A so-far little explored regulatory mechanism of the microtubule cytoskeleton, the posttranslational modifications of tubulin, emerge as candidate mechanisms involved in neuronal dysfunction, and thus, degeneration. Here we review the role of tubulin polyglutamylation, a prominent modification of neuronal microtubules. We discuss the current understanding of how polyglutamylation controls microtubule functions in healthy neurons, and how deregulation of this modification leads to neurodegeneration in mice and humans.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France.
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France.
| |
Collapse
|
57
|
Roles and mechanisms of BAP1 deubiquitinase in tumor suppression. Cell Death Differ 2021; 28:606-625. [PMID: 33462414 DOI: 10.1038/s41418-020-00709-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
The BAP1 gene has emerged as a major tumor suppressor mutated with various frequencies in numerous human malignancies, including uveal melanoma, malignant pleural mesothelioma, clear cell renal cell carcinoma, intrahepatic cholangiocarcinoma, hepatocellular carcinoma, and thymic epithelial tumors. BAP1 mutations are also observed at low frequency in other malignancies including breast, colorectal, pancreatic, and bladder cancers. BAP1 germline mutations are associated with high incidence of mesothelioma, uveal melanoma, and other cancers, defining the "BAP1 cancer syndrome." Interestingly, germline BAP1 mutations constitute an important paradigm for gene-environment interactions, as loss of BAP1 predisposes to carcinogen-induced tumorigenesis. Inactivating mutations of BAP1 are also identified in sporadic cancers, denoting the importance of this gene for normal tissue homeostasis and tumor suppression, although some oncogenic properties have also been attributed to BAP1. BAP1 belongs to the deubiquitinase superfamily of enzymes, which are responsible for the maturation and turnover of ubiquitin as well as the reversal of substrate ubiquitination, thus regulating ubiquitin signaling. BAP1 is predominantly nuclear and interacts with several chromatin-associated factors, assembling multi-protein complexes with mutually exclusive partners. BAP1 exerts its function through highly regulated deubiquitination of its substrates. As such, BAP1 orchestrates chromatin-associated processes including gene expression, DNA replication, and DNA repair. BAP1 also exerts cytoplasmic functions, notably in regulating Ca2+ signaling at the endoplasmic reticulum. This DUB is also subjected to multiple post-translational modifications, notably phosphorylation and ubiquitination, indicating that several signaling pathways tightly regulate its function. Recent progress indicated that BAP1 plays essential roles in multiple cellular processes including cell proliferation and differentiation, cell metabolism, as well as cell survival and death. In this review, we summarize the biological and molecular functions of BAP1 and explain how the inactivation of this DUB might cause human cancers. We also highlight some of the unresolved questions and suggest potential new directions.
Collapse
|
58
|
Li Z, Hajian C, Greene WC. Identification of unrecognized host factors promoting HIV-1 latency. PLoS Pathog 2020; 16:e1009055. [PMID: 33270809 PMCID: PMC7714144 DOI: 10.1371/journal.ppat.1009055] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022] Open
Abstract
To counter HIV latency, it is important to develop a better understanding of the full range of host factors promoting latency. Their identification could suggest new strategies to reactivate latent proviruses and subsequently kill the host cells (“shock and kill”), or to permanently silence these latent proviruses (“block and lock”). We recently developed a screening strategy termed “Reiterative Enrichment and Authentication of CRISPRi Targets” (REACT) that can unambiguously identify host genes promoting HIV latency, even in the presence of high background “noise” produced by the stochastic nature of HIV reactivation. After applying this strategy in four cell lines displaying different levels of HIV inducibility, we identified FTSJ3, TMEM178A, NICN1 and the Integrator Complex as host genes promoting HIV latency. shRNA knockdown of these four repressive factors significantly enhances HIV expression in primary CD4 T cells, and active HIV infection is preferentially found in cells expressing lower levels of these four factors. Mechanistically, we found that downregulation of these newly identified host inhibitors stimulates different stages of RNA Polymerase II-mediated transcription of HIV-1. The identification and validation of these new host inhibitors provide insight into the novel mechanisms that maintain HIV latency even when cells are activated and undergo cell division. The presence of a pool of latent HIV proviruses currently thwarts a cure for HIV-infected individuals. This “latent reservoir” is primarily composed of CD4 T cells that are infected with HIV but are indistinguishable from an uninfected T cell due to a lack of viral gene expression even when the cells are activated and undergo proliferation. This finding suggests there are powerful cellular mechanisms that hold HIV transcription in check even in stimulated cells allowing latent proviruses to remain hidden. Our goal was to identify and characterize these “unknown cellular factors”. We conducted genome-wide CRISPRi screens in multiple latently infected cell lines where each cell line displayed a different depth of latency as assessed by responsiveness to latency reversing agents. Application of our recently developed iterative screening strategy (REACT) allowed us to unambiguously identify and confirm four host factors that promote HIV latency. The latency promoting activity of these four factors (FTSJ3, TMEM178A, NICN1 and the Integrator Complex) were further validated in primary CD4 T cells, where their knockdown by shRNA significantly enhances latent HIV reactivation. In addition, we found that HIV infection preferentially occurs in cells expressing lower levels of these four factors. Mechanistically, our findings suggest that the newly identified host inhibitors likely block HIV transcription through different mechanisms. The identification and validation of these host inhibitors provides important new insights into how latency is maintained in T cells that could be useful for either activating and eliminating latently infected cells (“shock and kill”), or permanently silencing the integrated latent proviruses (“block and lock”).
Collapse
Affiliation(s)
- Zichong Li
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology, San Francisco, California, United States of America
| | - Cyrus Hajian
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology, San Francisco, California, United States of America
- Santa Rosa Junior College, Santa Rosa, California, United States of America
| | - Warner C. Greene
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology, San Francisco, California, United States of America
- Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
59
|
Lopes D, Maiato H. The Tubulin Code in Mitosis and Cancer. Cells 2020; 9:cells9112356. [PMID: 33114575 PMCID: PMC7692294 DOI: 10.3390/cells9112356] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/23/2022] Open
Abstract
The “tubulin code” combines different α/β-tubulin isotypes with several post-translational modifications (PTMs) to generate microtubule diversity in cells. During cell division, specific microtubule populations in the mitotic spindle are differentially modified, but only recently, the functional significance of the tubulin code, with particular emphasis on the role specified by tubulin PTMs, started to be elucidated. This is the case of α-tubulin detyrosination, which was shown to guide chromosomes during congression to the metaphase plate and allow the discrimination of mitotic errors, whose correction is required to prevent chromosomal instability—a hallmark of human cancers implicated in tumor evolution and metastasis. Although alterations in the expression of certain tubulin isotypes and associated PTMs have been reported in human cancers, it remains unclear whether and how the tubulin code has any functional implications for cancer cell properties. Here, we review the role of the tubulin code in chromosome segregation during mitosis and how it impacts cancer cell properties. In this context, we discuss the existence of an emerging “cancer tubulin code” and the respective implications for diagnostic, prognostic and therapeutic purposes.
Collapse
Affiliation(s)
- Danilo Lopes
- Chromosome Instability & Dynamics Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal;
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Helder Maiato
- Chromosome Instability & Dynamics Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal;
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Cell Division Group, Experimental Biology Unit, Department of Biomedicine, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Correspondence: ; Tel.: +351-22-040-8800
| |
Collapse
|
60
|
Power KM, Akella JS, Gu A, Walsh JD, Bellotti S, Morash M, Zhang W, Ramadan YH, Ross N, Golden A, Smith HE, Barr MM, O’Hagan R. Mutation of NEKL-4/NEK10 and TTLL genes suppress neuronal ciliary degeneration caused by loss of CCPP-1 deglutamylase function. PLoS Genet 2020; 16:e1009052. [PMID: 33064774 PMCID: PMC7592914 DOI: 10.1371/journal.pgen.1009052] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/28/2020] [Accepted: 08/14/2020] [Indexed: 12/29/2022] Open
Abstract
Ciliary microtubules are subject to post-translational modifications that act as a "Tubulin Code" to regulate motor traffic, binding proteins and stability. In humans, loss of CCP1, a cytosolic carboxypeptidase and tubulin deglutamylating enzyme, causes infantile-onset neurodegeneration. In C. elegans, mutations in ccpp-1, the homolog of CCP1, result in progressive degeneration of neuronal cilia and loss of neuronal function. To identify genes that regulate microtubule glutamylation and ciliary integrity, we performed a forward genetic screen for suppressors of ciliary degeneration in ccpp-1 mutants. We isolated the ttll-5(my38) suppressor, a mutation in a tubulin tyrosine ligase-like glutamylase gene. We show that mutation in the ttll-4, ttll-5, or ttll-11 gene suppressed the hyperglutamylation-induced loss of ciliary dye filling and kinesin-2 mislocalization in ccpp-1 cilia. We also identified the nekl-4(my31) suppressor, an allele affecting the NIMA (Never in Mitosis A)-related kinase NEKL-4/NEK10. In humans, NEK10 mutation causes bronchiectasis, an airway and mucociliary transport disorder caused by defective motile cilia. C. elegans NEKL-4 localizes to the ciliary base but does not localize to cilia, suggesting an indirect role in ciliary processes. This work defines a pathway in which glutamylation, a component of the Tubulin Code, is written by TTLL-4, TTLL-5, and TTLL-11; is erased by CCPP-1; is read by ciliary kinesins; and its downstream effects are modulated by NEKL-4 activity. Identification of regulators of microtubule glutamylation in diverse cellular contexts is important to the development of effective therapies for disorders characterized by changes in microtubule glutamylation. By identifying C. elegans genes important for neuronal and ciliary stability, our work may inform research into the roles of the tubulin code in human ciliopathies and neurodegenerative diseases.
Collapse
Affiliation(s)
- Kade M. Power
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Jyothi S. Akella
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Amanda Gu
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Jonathon D. Walsh
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Sebastian Bellotti
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Margaret Morash
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Winnie Zhang
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Yasmin H. Ramadan
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Nicole Ross
- Biology Department, Montclair State University, Montclair, NJ, United States of America
| | - Andy Golden
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Harold E. Smith
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maureen M. Barr
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Robert O’Hagan
- Biology Department, Montclair State University, Montclair, NJ, United States of America
| |
Collapse
|
61
|
Tubulin modifying enzymes as target for the treatment oftau-related diseases. Pharmacol Ther 2020; 218:107681. [PMID: 32961263 DOI: 10.1016/j.pharmthera.2020.107681] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/09/2020] [Indexed: 01/17/2023]
Abstract
In the brain of patients with Alzheimer's disease (AD), the number and length of microtubules (MTs) are significantly and selectively reduced. MTs are involved in a wide range of cellular functions, and defects of the microtubular system have emerged as a unifying hypothesis for the heterogeneous and variable clinical presentations of AD. MTs orchestrate their numerous functions through the spatiotemporal regulation of the binding of specialised microtubule-associated proteins (MAPs) and molecular motors. Covalent posttranslational modifications (PTMs) on the tubulin C-termini that protrude at the surface of MTs regulate the binding of these effectors. In neurons, MAP tau is highly abundant and its abnormal dissociation from MTs in the axon, cellular mislocalization and hyperphosphorylation, are primary events leading to neuronal death. Consequently, compounds targeting tau phosphorylation or aggregation are currently evaluated but their clinical significance has not been demonstrated yet. In this review, we discuss the emerging link between tubulin PTMs and tau dysfunction. In neurons, high levels of glutamylation and detyrosination profoundly impact the physicochemical properties at the surface of MTs. Moreover, in patients with early-onset progressive neurodegeneration, deleterious mutations in enzymes involved in modifying MTs at the surface have recently been identified, underscoring the importance of this enzymatic machinery in neurology. We postulate that pharmacologically targeting the tubulin-modifying enzymes holds promise as therapeutic approach for the treatment of neurodegenerative diseases.
Collapse
|
62
|
Jentzsch J, Sabri A, Speckner K, Lallinger-Kube G, Weiss M, Ersfeld K. Microtubule polyglutamylation is important for regulating cytoskeletal architecture and motility in Trypanosoma brucei. J Cell Sci 2020; 133:jcs248047. [PMID: 32843576 DOI: 10.1242/jcs.248047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/30/2020] [Indexed: 11/20/2022] Open
Abstract
The shape of kinetoplastids, such as Trypanosoma brucei, is precisely defined during the stages of the life cycle and governed by a stable subpellicular microtubule cytoskeleton. During the cell cycle and transitions between life cycle stages, this stability has to transiently give way to a dynamic behaviour to enable cell division and morphological rearrangements. How these opposing requirements of the cytoskeleton are regulated is poorly understood. Two possible levels of regulation are activities of cytoskeleton-associated proteins and microtubule post-translational modifications (PTMs). Here, we investigate the functions of two putative tubulin polyglutamylases in T. brucei, TTLL6A and TTLL12B. Depletion of both proteins leads to a reduction in tubulin polyglutamylation in situ and is associated with disintegration of the posterior cell pole, loss of the microtubule plus-end-binding protein EB1 and alterations of microtubule dynamics. We also observe a reduced polyglutamylation of the flagellar axoneme. Quantitative motility analysis reveals that the PTM imbalance correlates with a transition from directional to diffusive cell movement. These data show that microtubule polyglutamylation has an important role in regulating cytoskeletal architecture and motility in the parasite T. bruceiThis article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jana Jentzsch
- Molecular Parasitology, Department of Biology, University of Bayreuth, Universitätsstr. 30, 95447 Bayreuth, Germany
| | - Adal Sabri
- Experimental Physics I, Department of Physics, University of Bayreuth, Universitätsstr. 30, 95447 Bayreuth, Germany
| | - Konstantin Speckner
- Experimental Physics I, Department of Physics, University of Bayreuth, Universitätsstr. 30, 95447 Bayreuth, Germany
| | - Gertrud Lallinger-Kube
- Molecular Parasitology, Department of Biology, University of Bayreuth, Universitätsstr. 30, 95447 Bayreuth, Germany
| | - Matthias Weiss
- Experimental Physics I, Department of Physics, University of Bayreuth, Universitätsstr. 30, 95447 Bayreuth, Germany
| | - Klaus Ersfeld
- Molecular Parasitology, Department of Biology, University of Bayreuth, Universitätsstr. 30, 95447 Bayreuth, Germany
| |
Collapse
|
63
|
Xiong Z, Xia P, Zhu X, Geng J, Wang S, Ye B, Qin X, Qu Y, He L, Fan D, Du Y, Tian Y, Fan Z. Glutamylation of deubiquitinase BAP1 controls self-renewal of hematopoietic stem cells and hematopoiesis. J Exp Med 2020; 217:jem.20190974. [PMID: 31699823 PMCID: PMC7041701 DOI: 10.1084/jem.20190974] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/27/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022] Open
Abstract
Xiong et al. show that CCP3 performs deglutamylation of BAP1 to stabilize BAP1, which eliminates H2AK119Ub from Hoxa1 promoter and initiates Hoxa1 expression, leading to enhanced HSC self-renewal. All hematopoietic lineages are derived from a limited pool of hematopoietic stem cells (HSCs). Although the mechanisms underlying HSC self-renewal have been extensively studied, little is known about the role of protein glutamylation and deglutamylation in hematopoiesis. Here, we show that carboxypeptidase CCP3 is most highly expressed in BM cells among CCP members. CCP3 deficiency impairs HSC self-renewal and hematopoiesis. Deubiquitinase BAP1 is a substrate for CCP3 in HSCs. BAP1 is glutamylated at Glu651 by TTLL5 and TTLL7, and BAP1-E651A mutation abrogates BAP1 glutamylation. BAP1 glutamylation accelerates its ubiquitination to trigger its degradation. CCP3 can remove glutamylation of BAP1 to promote its stability, which enhances Hoxa1 expression, leading to HSC self-renewal. Bap1E651A mice produce higher numbers of LT-HSCs and peripheral blood cells. Moreover, TTLL5 and TTLL7 deficiencies sustain BAP1 stability to promote HSC self-renewal and hematopoiesis. Therefore, glutamylation and deglutamylation of BAP1 modulate HSC self-renewal and hematopoiesis.
Collapse
Affiliation(s)
- Zhen Xiong
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Pengyan Xia
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaoxiao Zhu
- Key Laboratory of RNA Biology of Chinese Academy of Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jingjing Geng
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuo Wang
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Buqing Ye
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiwen Qin
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Qu
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Luyun He
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Dongdong Fan
- Key Laboratory of RNA Biology of Chinese Academy of Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ying Du
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yong Tian
- University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of RNA Biology of Chinese Academy of Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zusen Fan
- Key Laboratory of Infection and Immunity of Chinese Academy of Sciences, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
64
|
Lopes AT, Hausrat TJ, Heisler FF, Gromova KV, Lombino FL, Fischer T, Ruschkies L, Breiden P, Thies E, Hermans-Borgmeyer I, Schweizer M, Schwarz JR, Lohr C, Kneussel M. Spastin depletion increases tubulin polyglutamylation and impairs kinesin-mediated neuronal transport, leading to working and associative memory deficits. PLoS Biol 2020; 18:e3000820. [PMID: 32866173 PMCID: PMC7485986 DOI: 10.1371/journal.pbio.3000820] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/11/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Mutations in the gene encoding the microtubule-severing protein spastin (spastic paraplegia 4 [SPG4]) cause hereditary spastic paraplegia (HSP), associated with neurodegeneration, spasticity, and motor impairment. Complicated forms (complicated HSP [cHSP]) further include cognitive deficits and dementia; however, the etiology and dysfunctional mechanisms of cHSP have remained unknown. Here, we report specific working and associative memory deficits upon spastin depletion in mice. Loss of spastin-mediated severing leads to reduced synapse numbers, accompanied by lower miniature excitatory postsynaptic current (mEPSC) frequencies. At the subcellular level, mutant neurons are characterized by longer microtubules with increased tubulin polyglutamylation levels. Notably, these conditions reduce kinesin-microtubule binding, impair the processivity of kinesin family protein (KIF) 5, and reduce the delivery of presynaptic vesicles and postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Rescue experiments confirm the specificity of these results by showing that wild-type spastin, but not the severing-deficient and disease-associated K388R mutant, normalizes the effects at the synaptic, microtubule, and transport levels. In addition, short hairpin RNA (shRNA)-mediated reduction of tubulin polyglutamylation on spastin knockout background normalizes KIF5 transport deficits and attenuates the loss of excitatory synapses. Our data provide a mechanism that connects spastin dysfunction with the regulation of kinesin-mediated cargo transport, synapse integrity, and cognition. This study identifies deficits in working and associative memory in spastin knockout mice, resembling the cognitive deficits described in humans with severe forms of SPG4-type hereditary spastic paraplegia. Mechanistically, the findings suggest that impaired microtubule growth, kinesin motility and cargo delivery of synaptic AMPA receptors may contribute to the etiology of the disease.
Collapse
Affiliation(s)
- André T. Lopes
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Torben J. Hausrat
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank F. Heisler
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kira V. Gromova
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franco L. Lombino
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Fischer
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Laura Ruschkies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Breiden
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edda Thies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Transgenic Animal Unit, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Morphology Unit, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jürgen R. Schwarz
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
65
|
Moutin MJ, Bosc C, Peris L, Andrieux A. Tubulin post-translational modifications control neuronal development and functions. Dev Neurobiol 2020; 81:253-272. [PMID: 33325152 PMCID: PMC8246997 DOI: 10.1002/dneu.22774] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/26/2020] [Accepted: 07/14/2020] [Indexed: 12/22/2022]
Abstract
Microtubules (MTs) are an essential component of the neuronal cytoskeleton; they are involved in various aspects of neuron development, maintenance, and functions including polarization, synaptic plasticity, and transport. Neuronal MTs are highly heterogeneous due to the presence of multiple tubulin isotypes and extensive post‐translational modifications (PTMs). These PTMs—most notably detyrosination, acetylation, and polyglutamylation—have emerged as important regulators of the neuronal microtubule cytoskeleton. With this review, we summarize what is currently known about the impact of tubulin PTMs on microtubule dynamics, neuronal differentiation, plasticity, and transport as well as on brain function in normal and pathological conditions, in particular during neuro‐degeneration. The main therapeutic approaches to neuro‐diseases based on the modulation of tubulin PTMs are also summarized. Overall, the review indicates how tubulin PTMs can generate a large number of functionally specialized microtubule sub‐networks, each of which is crucial to specific neuronal features.
Collapse
Affiliation(s)
- Marie-Jo Moutin
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble, France
| | - Christophe Bosc
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble, France
| | - Leticia Peris
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble, France
| | - Annie Andrieux
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble, France
| |
Collapse
|
66
|
The emerging role of tubulin posttranslational modifications in cilia and ciliopathies. BIOPHYSICS REPORTS 2020. [DOI: 10.1007/s41048-020-00111-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
67
|
Structural basis for polyglutamate chain initiation and elongation by TTLL family enzymes. Nat Struct Mol Biol 2020; 27:802-813. [PMID: 32747782 DOI: 10.1038/s41594-020-0462-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/12/2020] [Indexed: 11/08/2022]
Abstract
Glutamylation, introduced by tubulin tyrosine ligase-like (TTLL) enzymes, is the most abundant modification of brain tubulin. Essential effector proteins read the tubulin glutamylation pattern, and its misregulation causes neurodegeneration. TTLL glutamylases post-translationally add glutamates to internal glutamates in tubulin carboxy-terminal tails (branch initiation, through an isopeptide bond), and additional glutamates can extend these (elongation). TTLLs are thought to specialize in initiation or elongation, but the mechanistic basis for regioselectivity is unknown. We present cocrystal structures of murine TTLL6 bound to tetrahedral intermediate analogs that delineate key active-site residues that make this enzyme an elongase. We show that TTLL4 is exclusively an initiase and, through combined structural and phylogenetic analyses, engineer TTLL6 into a branch-initiating enzyme. TTLL glycylases add glycines post-translationally to internal glutamates, and we find that the same active-site residues discriminate between initiase and elongase glycylases. These active-site specializations of TTLL glutamylases and glycylases ultimately yield the chemical complexity of cellular microtubules.
Collapse
|
68
|
Lan H, Suzuki H, Nagatake T, Hosomi K, Ikegami K, Setou M, Kunisawa J. Impaired mucociliary motility enhances antigen-specific nasal IgA immune responses to a cholera toxin-based nasal vaccine. Int Immunol 2020; 32:559-568. [PMID: 32347929 PMCID: PMC9262165 DOI: 10.1093/intimm/dxaa029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 04/24/2020] [Indexed: 11/12/2022] Open
Abstract
Nasal mucosal tissues are equipped with physical barriers, mucus and cilia, on their surface. The mucus layer captures inhaled materials, and the cilia remove the inhaled materials from the epithelial layer by asymmetrical beating. The effect of nasal physical barriers on the vaccine efficacy remains to be investigated. Tubulin tyrosine ligase-like family, member 1 (Ttll1) is an essential enzyme for appropriate movement of the cilia on respiratory epithelium, and its deficiency (Ttll1-KO) leads to mucus accumulation in the nasal cavity. Here, when mice were intra-nasally immunized with pneumococcal surface protein A (PspA, as vaccine antigen) together with cholera toxin (CT, as mucosal adjuvant), Ttll1-KO mice showed higher levels of PspA-specific IgA in the nasal wash and increased numbers of PspA-specific IgA-producing plasma cells in the nasal passages when compared with Ttll1 hetero (He) mice. Mucus removal by N-acetylcysteine did not affect the enhanced immune responses in Ttll1-KO mice versus Ttll1-He mice. Immunohistological and flow cytometry analyses revealed that retention time of PspA in the nasal cavity in Ttll1-KO mice was longer than that in Ttll1-He mice. Consistently, uptake of PspA by dendritic cells was higher in the nasopharynx-associated lymphoid tissue (NALT) of Ttll1-KO mice than that of Ttll1-He mice. These results indicate that the ciliary function of removing vaccine antigen from the NALT epithelial layer is a critical determinant of the efficacy of nasal vaccine.
Collapse
Affiliation(s)
- Huangwenxian Lan
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hidehiko Suzuki
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Koji Ikegami
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mitsutoshi Setou
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, Kobe University, Hyogo, Japan
- Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
69
|
Roll-Mecak A. The Tubulin Code in Microtubule Dynamics and Information Encoding. Dev Cell 2020; 54:7-20. [PMID: 32634400 PMCID: PMC11042690 DOI: 10.1016/j.devcel.2020.06.008] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 05/08/2020] [Accepted: 06/03/2020] [Indexed: 01/05/2023]
Abstract
Microtubules are non-covalent mesoscale polymers central to the eukaryotic cytoskeleton. Microtubule structure, dynamics, and mechanics are modulated by a cell's choice of tubulin isoforms and post-translational modifications, a "tubulin code," which is thought to support the diverse morphology and dynamics of microtubule arrays across various cell types, cell cycle, and developmental stages. We give a brief historical overview of research into tubulin diversity and highlight recent progress toward uncovering the mechanistic underpinnings of the tubulin code. As a large number of essential pathways converge upon the microtubule cytoskeleton, understanding how cells utilize tubulin diversity is crucial to understanding cellular physiology and disease.
Collapse
Affiliation(s)
- Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA; Biochemistry and Biophysics Center, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
70
|
Amargant F, Barragan M, Vassena R, Vernos I. Insights of the tubulin code in gametes and embryos: from basic research to potential clinical applications in humans†. Biol Reprod 2020; 100:575-589. [PMID: 30247519 DOI: 10.1093/biolre/ioy203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 07/05/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
Microtubules are intracellular filaments that define in space and in time a large number of essential cellular functions such as cell division, morphology and motility, intracellular transport and flagella and cilia assembly. They are therefore essential for spermatozoon and oocyte maturation and function, and for embryo development. The dynamic and functional properties of the microtubules are in large part defined by various classes of interacting proteins including MAPs (microtubule associated proteins), microtubule-dependent motors, and severing and modifying enzymes. Multiple mechanisms regulate these interactions. One of them is defined by the high diversity of the microtubules themselves generated by the combination of different tubulin isotypes and by several tubulin post-translational modifications (PTMs). This generates a so-called tubulin code that finely regulates the specific set of proteins that associates with a given microtubule thereby defining the properties and functions of the network. Here we provide an in depth review of the current knowledge on the tubulin isotypes and PTMs in spermatozoa, oocytes, and preimplantation embryos in various model systems and in the human species. We focus on functional implications of the tubulin code for cytoskeletal function, particularly in the field of human reproduction and development, with special emphasis on gamete quality and infertility. Finally, we discuss some of the knowledge gaps and propose future research directions.
Collapse
Affiliation(s)
- Farners Amargant
- Clínica EUGIN, Barcelona, Spain.,Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | | | - Isabelle Vernos
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
71
|
Chin HG, Esteve PO, Ruse C, Lee J, Schaus SE, Pradhan S, Hansen U. The microtubule-associated histone methyltransferase SET8, facilitated by transcription factor LSF, methylates α-tubulin. J Biol Chem 2020; 295:4748-4759. [PMID: 32111740 PMCID: PMC7135998 DOI: 10.1074/jbc.ra119.010951] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/21/2020] [Indexed: 01/20/2023] Open
Abstract
Microtubules are cytoskeletal structures critical for mitosis, cell motility, and protein and organelle transport and are a validated target for anticancer drugs. However, how tubulins are regulated and recruited to support these distinct cellular processes is incompletely understood. Posttranslational modifications of tubulins are proposed to regulate microtubule function and dynamics. Although many of these modifications have been investigated, only one prior study reports tubulin methylation and an enzyme responsible for this methylation. Here we used in vitro radiolabeling, MS, and immunoblotting approaches to monitor protein methylation and immunoprecipitation, immunofluorescence, and pulldown approaches to measure protein-protein interactions. We demonstrate that N-lysine methyltransferase 5A (KMT5A or SET8/PR-Set7), which methylates lysine 20 in histone H4, bound α-tubulin and methylated it at a specific lysine residue, Lys311 Furthermore, late SV40 factor (LSF)/CP2, a known transcription factor, bound both α-tubulin and SET8 and enhanced SET8-mediated α-tubulin methylation in vitro In addition, we found that the ability of LSF to facilitate this methylation is countered by factor quinolinone inhibitor 1 (FQI1), a specific small-molecule inhibitor of LSF. These findings suggest the general model that microtubule-associated proteins, including transcription factors, recruit or stimulate protein-modifying enzymes to target tubulins. Moreover, our results point to dual functions for SET8 and LSF not only in chromatin regulation but also in cytoskeletal modification.
Collapse
Affiliation(s)
- Hang Gyeong Chin
- New England Biolabs, Ipswich, Massachusetts 01938
- MCBB Graduate Program, Graduate School of Arts and Sciences, Boston University, Boston, Massachusetts 02215
| | | | | | - Jiyoung Lee
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Ipsin-gil, Jeongeup-si, Jeollabuk-do 56212, South Korea
| | - Scott E Schaus
- Center for Molecular Discovery, Boston University, Boston, Massachusetts 02215
| | | | - Ulla Hansen
- MCBB Graduate Program, Graduate School of Arts and Sciences, Boston University, Boston, Massachusetts 02215
- Department of Biology, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
72
|
Shin SC, Im SK, Jang EH, Jin KS, Hur EM, Kim EE. Structural and Molecular Basis for Katanin-Mediated Severing of Glutamylated Microtubules. Cell Rep 2020; 26:1357-1367.e5. [PMID: 30699360 DOI: 10.1016/j.celrep.2019.01.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/09/2018] [Accepted: 01/04/2019] [Indexed: 12/28/2022] Open
Abstract
Katanin was the first microtubule (MT)-severing enzyme discovered, but how katanin executes MT severing remains poorly understood. Here, we report X-ray crystal structures of the apo and ATPγS-bound states of the catalytic AAA domain of human katanin p60 at 3.0 and 2.9 Å resolution, respectively. Comparison of the two structures reveals conformational changes induced by ATP binding and how such changes ensure hexamer stability. Moreover, we uncover structural details of pore loops (PLs) and show that Arg283, a residue unique to katanin among MT-severing enzymes, protrudes from PL1 and lines the entry of the catalytic pore. Functional studies suggest that PL1 and Arg283 play essential roles in the recognition and remodeling of the glutamylated, C-terminal tubulin tail and regulation of axon growth. In addition, domain-swapping experiments in katanin and spastin suggest that the non-homologous N-terminal region, which contains the MT-interacting and trafficking domain and a linker, confers specificity to the severing process.
Collapse
Affiliation(s)
- Sang Chul Shin
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sun-Kyoung Im
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Eun-Hae Jang
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; Department of Neuroscience, Korea University of Science and Technology, Daejeon, Republic of Korea; Department of Neuroscience, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyeong Sik Jin
- Pohang Accelerator Laboratory, Pohang University of Science and Technology, Pohang, Kyungbuk, 37673, Republic of Korea
| | - Eun-Mi Hur
- Department of Neuroscience, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Eunice EunKyeong Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
73
|
The tubulin code and its role in controlling microtubule properties and functions. Nat Rev Mol Cell Biol 2020; 21:307-326. [PMID: 32107477 DOI: 10.1038/s41580-020-0214-3] [Citation(s) in RCA: 421] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Microtubules are core components of the eukaryotic cytoskeleton with essential roles in cell division, shaping, motility and intracellular transport. Despite their functional heterogeneity, microtubules have a highly conserved structure made from almost identical molecular building blocks: the tubulin proteins. Alternative tubulin isotypes and a variety of post-translational modifications control the properties and functions of the microtubule cytoskeleton, a concept known as the 'tubulin code'. Here we review the current understanding of the molecular components of the tubulin code and how they impact microtubule properties and functions. We discuss how tubulin isotypes and post-translational modifications control microtubule behaviour at the molecular level and how this translates into physiological functions at the cellular and organism levels. We then go on to show how fine-tuning of microtubule function by some tubulin modifications can affect homeostasis and how perturbation of this fine-tuning can lead to a range of dysfunctions, many of which are linked to human disease.
Collapse
|
74
|
Bodakuntla S, Schnitzler A, Villablanca C, Gonzalez-Billault C, Bieche I, Janke C, Magiera MM. Tubulin polyglutamylation is a general traffic-control mechanism in hippocampal neurons. J Cell Sci 2020; 133:jcs241802. [PMID: 31932508 DOI: 10.1242/jcs.241802] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/23/2019] [Indexed: 08/31/2023] Open
Abstract
Neurons are highly complex cells that heavily rely on intracellular transport to distribute a range of functionally essential cargoes within the cell. Post-translational modifications of tubulin are emerging as mechanisms for regulating microtubule functions, but their impact on neuronal transport is only marginally understood. Here, we have systematically studied the impact of post-translational polyglutamylation on axonal transport. In cultured hippocampal neurons, deletion of a single deglutamylase, CCP1 (also known as AGTPBP1), is sufficient to induce abnormal accumulation of polyglutamylation, i.e. hyperglutamylation. We next investigated how hyperglutamylation affects axonal transport of a range of functionally different neuronal cargoes: mitochondria, lysosomes, LAMP1 endosomes and BDNF vesicles. Strikingly, we found a reduced motility for all these cargoes, suggesting that polyglutamylation could act as a regulator of cargo transport in neurons. This, together with the recent discovery that hyperglutamylation induces neurodegeneration, makes it likely that perturbed neuronal trafficking could be one of the central molecular causes underlying this novel type of degeneration.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Anne Schnitzler
- Institut Curie, PSL Research University, Department of Genetics, F-75005 Paris, France
| | - Cristopher Villablanca
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile
- Department of Biology, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Christian Gonzalez-Billault
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile
- Department of Biology, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Ivan Bieche
- Institut Curie, PSL Research University, Department of Genetics, F-75005 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, F-75005 Paris, 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
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| |
Collapse
|
75
|
Joachimiak E, Waclawek E, Niziolek M, Osinka A, Fabczak H, Gaertig J, Wloga D. The LisH Domain-Containing N-Terminal Fragment is Important for the Localization, Dimerization, and Stability of Katnal2 in Tetrahymena. Cells 2020; 9:cells9020292. [PMID: 31991798 PMCID: PMC7072489 DOI: 10.3390/cells9020292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 12/12/2022] Open
Abstract
Katanin-like 2 protein (Katnal2) orthologs have a tripartite domain organization. Two highly conserved regions, an N-terminal LisH (Lis-homology) domain and a C-terminal AAA catalytic domain, are separated by a less conserved linker. The AAA domain of Katnal2 shares the highest amino acid sequence homology with the AAA domain of the canonical katanin p60. Katnal2 orthologs are present in a wide range of eukaryotes, from protists to humans. In the ciliate Tetrahymena thermophila, a Katnal2 ortholog, Kat2, co-localizes with the microtubular structures, including basal bodies and ciliary outer doublets, and this co-localization is sensitive to levels of microtubule glutamylation. The functional analysis of Kat2 domains suggests that an N-terminal fragment containing a LisH domain plays a role in the subcellular localization, dimerization, and stability of Kat2.
Collapse
Affiliation(s)
- Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology PAS, 3 Pasteur, 02-093 Warsaw, Poland; (E.J.); (E.W.); (M.N.); (A.O.); (H.F.)
| | - Ewa Waclawek
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology PAS, 3 Pasteur, 02-093 Warsaw, Poland; (E.J.); (E.W.); (M.N.); (A.O.); (H.F.)
| | - Michal Niziolek
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology PAS, 3 Pasteur, 02-093 Warsaw, Poland; (E.J.); (E.W.); (M.N.); (A.O.); (H.F.)
| | - Anna Osinka
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology PAS, 3 Pasteur, 02-093 Warsaw, Poland; (E.J.); (E.W.); (M.N.); (A.O.); (H.F.)
| | - Hanna Fabczak
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology PAS, 3 Pasteur, 02-093 Warsaw, Poland; (E.J.); (E.W.); (M.N.); (A.O.); (H.F.)
| | - Jacek Gaertig
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA;
| | - Dorota Wloga
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology PAS, 3 Pasteur, 02-093 Warsaw, Poland; (E.J.); (E.W.); (M.N.); (A.O.); (H.F.)
- Correspondence: ; Tel.: +48-(22)-5892338
| |
Collapse
|
76
|
Bodakuntla S, Janke C, Magiera MM. Knocking Out Multiple Genes in Cultured Primary Neurons to Study Tubulin Posttranslational Modifications. Methods Mol Biol 2020; 2101:327-351. [PMID: 31879912 DOI: 10.1007/978-1-0716-0219-5_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Microtubules, as integral part of the eukaryotic cytoskeleton, exert numerous essential functions in cells. A mechanism to control these diverse functions are the posttranslational modifications of tubulin. Despite being known for decades, relatively little insight into the cellular functions of these modifications has been gained so far. The discovery of tubulin-modifying enzymes and a growing number of available knockout mice now allow working with primary cells from those mouse models to address biological functions and molecular mechanisms behind those modifications. However, a number of those mouse models show either lethality or sterility, making it difficult to impossible to obtain a sufficient number of animals for a systematic study with primary cells. Moreover, many of those modifications are controlled by several redundant enzymes, and it is often necessary to knock out several enzymes in parallel to obtain a significant change in a given tubulin modification. Here we describe a method to generate primary cells with combinatorial knockout genotypes using conditional knockout mice. The conditional alleles are converted into knockout in the cultured primary cells by transduction with a lentivirus encoding cre-recombinase. This approach has allowed us to knock out the two main brain deglutamylases in mouse primary neurons, which leads to strongly increased polyglutamylation in these cells. Our method can be applied to measure different cellular processes, such as axonal transport, for which it can be combined with the expression of different fluorescent reporters to label intracellular proteins. Using a panel of conditional knockout mice, our method can further be applied to study the functions of a variety of tubulin modifications that require simultaneous knockout of multiple genes.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France.
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France.
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| |
Collapse
|
77
|
Bodakuntla S, Magiera MM, Janke C. Measuring the Impact of Tubulin Posttranslational Modifications on Axonal Transport. Methods Mol Biol 2020; 2101:353-370. [PMID: 31879913 DOI: 10.1007/978-1-0716-0219-5_20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Axonal transport is a process essential for neuronal function and survival that takes place on the cellular highways-the microtubules. It requires three major components: the microtubules that serve as tracks for the transport, the motor proteins that drive the movement, and the transported cargoes with their adaptor proteins. Axonal transport could be controlled by tubulin posttranslational modifications, which by decorating specific microtubule tracks could determine the specificity of cargo delivery inside neurons. However, it appears that the effects of tubulin modifications on transport can be rather subtle, and might thus be easily overlooked depending on which parameter of the transport process is analyzed. Here we propose an analysis paradigm that allows detecting rather subtle alterations in neuronal transport, as induced for instance by accumulation of posttranslational polyglutamylation. Analyzing mitochondria movements in axons, we found that neither the average speed nor the distance traveled were affected by hyperglutamylation, but we detected an about 50% reduction of the overall motility, suggesting that polyglutamylation controls the efficiency of mitochondria transport in axons. Our protocol can readily be expanded to the analysis of the impact of other tubulin modifications on the transport of a range of different neuronal cargoes.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France. .,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France. .,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| |
Collapse
|
78
|
Bompard G, van Dijk J, Cau J, Lannay Y, Marcellin G, Lawera A, van der Laan S, Rogowski K. CSAP Acts as a Regulator of TTLL-Mediated Microtubule Glutamylation. Cell Rep 2019; 25:2866-2877.e5. [PMID: 30517872 DOI: 10.1016/j.celrep.2018.10.095] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/22/2017] [Accepted: 10/25/2018] [Indexed: 01/22/2023] Open
Abstract
Tubulin glutamylation is a reversible posttranslational modification that accumulates on stable microtubules (MTs). While abnormally high levels of this modification lead to a number of disorders such as male sterility, retinal degeneration, and neurodegeneration, very little is known about the molecular mechanisms underlying the regulation of glutamylase activity. Here, we found that CSAP forms a complex with TTLL5, and we demonstrate that the two proteins regulate their reciprocal abundance. Moreover, we show that CSAP increases TTLL5-mediated glutamylation and identify the TTLL5-interacting domain. Deletion of this domain leads to complete loss of CSAP activating function without impacting its MT binding. Binding of CSAP to TTLL5 promotes relocalization of TTLL5 toward MTs. Finally, we show that CSAP binds and activates all of the remaining autonomously active TTLL glutamylases. As such, we present CSAP as a major regulator of tubulin glutamylation and associated functions.
Collapse
Affiliation(s)
- Guillaume Bompard
- Institute of Human Genetics (IGH), UMR9002 CNRS-University of Montpellier, 34094 Cedex 5, 141 Rue de la Cardonille, 34090 Montpellier, France
| | - Juliette van Dijk
- Institute of Human Genetics (IGH), UMR9002 CNRS-University of Montpellier, 34094 Cedex 5, 141 Rue de la Cardonille, 34090 Montpellier, France
| | - Julien Cau
- Institute of Human Genetics (IGH), UMR9002 CNRS-University of Montpellier, 34094 Cedex 5, 141 Rue de la Cardonille, 34090 Montpellier, France
| | - Yoann Lannay
- Institute of Human Genetics (IGH), UMR9002 CNRS-University of Montpellier, 34094 Cedex 5, 141 Rue de la Cardonille, 34090 Montpellier, France
| | - Guillaume Marcellin
- Institute of Human Genetics (IGH), UMR9002 CNRS-University of Montpellier, 34094 Cedex 5, 141 Rue de la Cardonille, 34090 Montpellier, France
| | - Aleksandra Lawera
- Institute of Human Genetics (IGH), UMR9002 CNRS-University of Montpellier, 34094 Cedex 5, 141 Rue de la Cardonille, 34090 Montpellier, France
| | - Siem van der Laan
- Institute of Human Genetics (IGH), UMR9002 CNRS-University of Montpellier, 34094 Cedex 5, 141 Rue de la Cardonille, 34090 Montpellier, France
| | - Krzysztof Rogowski
- Institute of Human Genetics (IGH), UMR9002 CNRS-University of Montpellier, 34094 Cedex 5, 141 Rue de la Cardonille, 34090 Montpellier, France.
| |
Collapse
|
79
|
Bayless BA, Navarro FM, Winey M. Motile Cilia: Innovation and Insight From Ciliate Model Organisms. Front Cell Dev Biol 2019; 7:265. [PMID: 31737631 PMCID: PMC6838636 DOI: 10.3389/fcell.2019.00265] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022] Open
Abstract
Ciliates are a powerful model organism for the study of basal bodies and motile cilia. These single-celled protists contain hundreds of cilia organized in an array making them an ideal system for both light and electron microscopy studies. Isolation and subsequent proteomic analysis of both cilia and basal bodies have been carried out to great success in ciliates. These studies reveal that ciliates share remarkable protein conservation with metazoans and have identified a number of essential basal body/ciliary proteins. Ciliates also boast a genetic and molecular toolbox that allows for facile manipulation of ciliary genes. Reverse genetics studies in ciliates have expanded our understanding of how cilia are positioned within an array, assembled, stabilized, and function at a molecular level. The advantages of cilia number coupled with a robust genetic and molecular toolbox have established ciliates as an ideal system for motile cilia and basal body research and prove a promising system for future research.
Collapse
Affiliation(s)
- Brian A Bayless
- Department of Biology, Santa Clara University, Santa Clara, CA, United States
| | - Francesca M Navarro
- Department of Biology, Santa Clara University, Santa Clara, CA, United States
| | - Mark Winey
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
80
|
Gardiner J. Posttranslational modification of plant microtubules. PLANT SIGNALING & BEHAVIOR 2019; 14:e1654818. [PMID: 31564233 PMCID: PMC6768230 DOI: 10.1080/15592324.2019.1654818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 06/10/2023]
Abstract
Microtubules in eukaryotes have a number of posttranslational modifications catalyzed by an array of enzymes. These modifications alter the properties of the microtubules and the ways in which they interact with partner proteins. In recent years many of the enzymes which modify the microtubules have been identified in animals and protozoans. Relatively little work has been done on their function in plants, however. This study uses bioinformatics to identify homologues of these enzymes in plant species from the green alga Chlamydomonas reiinhardtii to the angiosperm Arabidopsis thaliana. Many are conserved and this gives insight into the likely future direction of this dynamic field.
Collapse
|
81
|
Shah P, Martinez M, Chakraborty M, Kota S, Shah B, Lee J, Peel N. Double mutants of TTLL glutamylase genes have little to no difference in viability or brood size. MICROPUBLICATION BIOLOGY 2019; 2019:10.17912/micropub.biology.000156. [PMID: 32550433 PMCID: PMC7252309 DOI: 10.17912/micropub.biology.000156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Pooja Shah
- The College of New Jersey, 2000 Pennington Rd, Ewing, NJ 08618, USA
| | - Monica Martinez
- The College of New Jersey, 2000 Pennington Rd, Ewing, NJ 08618, USA
| | | | - Sharwani Kota
- The College of New Jersey, 2000 Pennington Rd, Ewing, NJ 08618, USA
| | - Bhumi Shah
- The College of New Jersey, 2000 Pennington Rd, Ewing, NJ 08618, USA
| | - Jessica Lee
- The College of New Jersey, 2000 Pennington Rd, Ewing, NJ 08618, USA
| | - Nina Peel
- The College of New Jersey, 2000 Pennington Rd, Ewing, NJ 08618, USA,
Correspondence to: Nina Peel ()
| |
Collapse
|
82
|
Junker AD, Soh AWJ, O'Toole ET, Meehl JB, Guha M, Winey M, Honts JE, Gaertig J, Pearson CG. Microtubule glycylation promotes attachment of basal bodies to the cell cortex. J Cell Sci 2019; 132:jcs.233726. [PMID: 31243050 DOI: 10.1242/jcs.233726] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022] Open
Abstract
Motile cilia generate directed hydrodynamic flow that is important for the motility of cells and extracellular fluids. To optimize directed hydrodynamic flow, motile cilia are organized and oriented into a polarized array. Basal bodies (BBs) nucleate and position motile cilia at the cell cortex. Cytoplasmic BB-associated microtubules are conserved structures that extend from BBs. By using the ciliate, Tetrahymena thermophila, combined with EM-tomography and light microscopy, we show that BB-appendage microtubules assemble coincidently with new BB assembly and that they are attached to the cell cortex. These BB-appendage microtubules are specifically marked by post translational modifications of tubulin, including glycylation. Mutations that prevent glycylation shorten BB-appendage microtubules and disrupt BB positioning and cortical attachment. Consistent with the attachment of BB-appendage microtubules to the cell cortex to position BBs, mutations that disrupt the cellular cortical cytoskeleton disrupt the cortical attachment and positioning of BBs. In summary, BB-appendage microtubules promote the organization of ciliary arrays through attachment to the cell cortex.
Collapse
Affiliation(s)
- Anthony D Junker
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Adam W J Soh
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Eileen T O'Toole
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80302, USA
| | - Janet B Meehl
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80302, USA
| | - Mayukh Guha
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Mark Winey
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Jerry E Honts
- Department of Biology, Drake University, 2507 University Avenue, Des Moines, IA 50311, USA
| | - Jacek Gaertig
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Chad G Pearson
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
83
|
Gheiratmand L, Coyaud E, Gupta GD, Laurent EMN, Hasegan M, Prosser SL, Gonçalves J, Raught B, Pelletier L. Spatial and proteomic profiling reveals centrosome-independent features of centriolar satellites. EMBO J 2019; 38:e101109. [PMID: 31304627 PMCID: PMC6627244 DOI: 10.15252/embj.2018101109] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 12/19/2022] Open
Abstract
Centriolar satellites are small electron-dense granules that cluster in the vicinity of centrosomes. Satellites have been implicated in multiple critical cellular functions including centriole duplication, centrosome maturation, and ciliogenesis, but their precise composition and assembly properties have remained poorly explored. Here, we perform in vivo proximity-dependent biotin identification (BioID) on 22 human satellite proteins, to identify 2,113 high-confidence interactions among 660 unique polypeptides. Mining this network, we validate six additional satellite components. Analysis of the satellite interactome, combined with subdiffraction imaging, reveals the existence of multiple unique microscopically resolvable satellite populations that display distinct protein interaction profiles. We further show that loss of satellites in PCM1-depleted cells results in a dramatic change in the satellite interaction landscape. Finally, we demonstrate that satellite composition is largely unaffected by centriole depletion or disruption of microtubules, indicating that satellite assembly is centrosome-independent. Together, our work offers the first systematic spatial and proteomic profiling of human centriolar satellites and paves the way for future studies aimed at better understanding the biogenesis and function(s) of these enigmatic structures.
Collapse
Affiliation(s)
- Ladan Gheiratmand
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
| | - Etienne Coyaud
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoONCanada
| | - Gagan D Gupta
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
- Present address:
Department of Chemistry and BiologyRyerson UniversityTorontoONCanada
| | | | - Monica Hasegan
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
| | - Suzanna L Prosser
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
| | - João Gonçalves
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
| | - Brian Raught
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoONCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
| | - Laurence Pelletier
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| |
Collapse
|
84
|
Basmaciyan L, Robinson DR, Azas N, Casanova M. (De)glutamylation and cell death in Leishmania parasites. PLoS Negl Trop Dis 2019; 13:e0007264. [PMID: 31017892 PMCID: PMC6502457 DOI: 10.1371/journal.pntd.0007264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/06/2019] [Accepted: 02/26/2019] [Indexed: 11/18/2022] Open
Abstract
Trypanosomatids are flagellated protozoan parasites that are very unusual in terms of cytoskeleton organization but also in terms of cell death. Most of the Trypanosomatid cytoskeleton consists of microtubules, forming different substructures including a subpellicular corset. Oddly, the actin network appears structurally and functionally different from other eukaryotic actins. And Trypanosomatids have an apoptotic phenotype under cell death conditions, but the pathways involved are devoid of key mammal proteins such as caspases or death receptors, and the triggers involved in apoptotic induction remain unknown. In this article, we have studied the role of the post-translational modifications, deglutamylation and polyglutamylation, in Leishmania. We have shown that Leishmania apoptosis was linked to polyglutamylation and hypothesized that the cell survival process autophagy was linked to deglutamylation. A balance seems to be established between polyglutamylation and deglutamylation, with imbalance inducing microtubule or other protein modifications characterizing either cell death if polyglutamylation was prioritized, or the cell survival process of autophagy if deglutamylation was prioritized. This emphasizes the role of post-translational modifications in cell biology, inducing cell death or cell survival of infectious agents. Leishmania are unique unicellular organisms in terms of cytoskeleton organization and mechanisms of cell death. For example, the major cytoskeletal components of these parasites are microtubules, which form a subpellicular corset. In terms of cell death, an apoptotic phenotype has been characterized in Leishmania but the pathways remain unknown, being devoid of key mammal cell death proteins. In a previous article, we demonstrated that the cytoskeleton of this parasite is extensively glutamylated but, paradoxically, overexpression or inhibition of polyglutamylase expression have limited visible cellular consequences. In this manuscript, we have highlighted the link between polyglutamylation and Leishmania cell death, suggesting the importance of the polyglutamylation/deglutamylation balance in this parasite. Further, we have identified, for the first time in Leishmania, deglutamylases, among which one that, in an original manner, deglutamylates glutamates at branching points but also long glutamate side chains. This work emphasizes the role of post-translational modifications as essential regulators of protein function, not only of mammal cells such as neurons or ciliated/flagellated cells, but also of infectious agents. This work suggests an important and discernible “live or die”—“cell death or autophagy” balance pathway and the conceptual mechanism that is involved in cellular decision making.
Collapse
Affiliation(s)
- Louise Basmaciyan
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| | | | - Nadine Azas
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| | - Magali Casanova
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- * E-mail:
| |
Collapse
|
85
|
Purification of tubulin with controlled post-translational modifications by polymerization–depolymerization cycles. Nat Protoc 2019; 14:1634-1660. [DOI: 10.1038/s41596-019-0153-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/22/2019] [Indexed: 12/27/2022]
|
86
|
Lessard DV, Zinder OJ, Hotta T, Verhey KJ, Ohi R, Berger CL. Polyglutamylation of tubulin's C-terminal tail controls pausing and motility of kinesin-3 family member KIF1A. J Biol Chem 2019; 294:6353-6363. [PMID: 30770469 DOI: 10.1074/jbc.ra118.005765] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/11/2019] [Indexed: 01/06/2023] Open
Abstract
The kinesin-3 family member KIF1A plays a critical role in site-specific neuronal cargo delivery during axonal transport. KIF1A cargo is mislocalized in many neurodegenerative diseases, indicating that KIF1A's highly efficient, superprocessive motility along axonal microtubules needs to be tightly regulated. One potential regulatory mechanism may be through posttranslational modifications (PTMs) of axonal microtubules. These PTMs often occur on the C-terminal tails of the microtubule tracks, act as molecular "traffic signals" helping to direct kinesin motor cargo delivery, and include C-terminal tail polyglutamylation important for KIF1A cargo transport. KIF1A initially interacts with microtubule C-terminal tails through its K-loop, a positively charged surface loop of the KIF1A motor domain. However, the role of the K-loop in KIF1A motility and response to perturbations in C-terminal tail polyglutamylation is underexplored. Using single-molecule imaging, we present evidence that KIF1A pauses on different microtubule lattice structures, linking multiple processive segments together and contributing to KIF1A's characteristic superprocessive run length. Furthermore, modifications of the KIF1A K-loop or tubulin C-terminal tail polyglutamylation reduced KIF1A pausing and overall run length. These results suggest a new mechanism to regulate KIF1A motility via pauses mediated by K-loop/polyglutamylated C-terminal tail interactions, providing further insight into KIF1A's role in axonal transport.
Collapse
Affiliation(s)
- Dominique V Lessard
- From the Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont 05405 and
| | - Oraya J Zinder
- From the Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont 05405 and
| | - Takashi Hotta
- the Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Kristen J Verhey
- the Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Ryoma Ohi
- the Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Christopher L Berger
- From the Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont 05405 and
| |
Collapse
|
87
|
Kubo T, Oda T. Chlamydomonas as a tool to study tubulin polyglutamylation. Microscopy (Oxf) 2019; 68:80-91. [PMID: 30364995 DOI: 10.1093/jmicro/dfy044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/11/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
The diversity of α- and β-tubulin is facilitated by various post-translational modifications (PTMs), such as acetylation, tyrosination, glycylation, glutamylation, phosphorylation and methylation. These PTMs affect the stability and structure of microtubules as well as the interaction between microtubules and microtubule-associated proteins, including molecular motors. Therefore, it is extremely important to investigate the roles of tubulin PTMs for understanding the cell cycle, cell motility and intracellular trafficking. Tubulin PTMs were first studied in the 1980s, and considerable progress has been made since then; it is likely that additional mechanisms remain yet to be elucidated. Here, we discuss one such modification, tubulin glutamylation, and introduce our research on the eukaryotic flagellum of the unicellular green alga Chlamydomonas reinhardtii.
Collapse
Affiliation(s)
- Tomohiro Kubo
- Department of Anatomy and Structural Biology, Graduate School of Medicine, University of Yamanashi, Shimokato, Chuo, Yamanashi, Japan
| | - Toshiyuki Oda
- Department of Anatomy and Structural Biology, Graduate School of Medicine, University of Yamanashi, Shimokato, Chuo, Yamanashi, Japan
| |
Collapse
|
88
|
Silva CG, Peyre E, Adhikari MH, Tielens S, Tanco S, Van Damme P, Magno L, Krusy N, Agirman G, Magiera MM, Kessaris N, Malgrange B, Andrieux A, Janke C, Nguyen L. Cell-Intrinsic Control of Interneuron Migration Drives Cortical Morphogenesis. Cell 2019; 172:1063-1078.e19. [PMID: 29474907 PMCID: PMC5847171 DOI: 10.1016/j.cell.2018.01.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/17/2017] [Accepted: 01/24/2018] [Indexed: 11/16/2022]
Abstract
Interneurons navigate along multiple tangential paths to settle into appropriate cortical layers. They undergo a saltatory migration paced by intermittent nuclear jumps whose regulation relies on interplay between extracellular cues and genetic-encoded information. It remains unclear how cycles of pause and movement are coordinated at the molecular level. Post-translational modification of proteins contributes to cell migration regulation. The present study uncovers that carboxypeptidase 1, which promotes post-translational protein deglutamylation, controls the pausing of migrating cortical interneurons. Moreover, we demonstrate that pausing during migration attenuates movement simultaneity at the population level, thereby controlling the flow of interneurons invading the cortex. Interfering with the regulation of pausing not only affects the size of the cortical interneuron cohort but also impairs the generation of age-matched projection neurons of the upper layers. CCP1 controls MLCK activity by processing its polyglutamate carboxy-terminal CCP1 controls the stereotypic two-stroke cycle of cortical interneuron migration Asynchronous pausing during interneuron migration controls cortical invasion Interneuron cortical invasion modulates generation of age-matched projection neurons
Collapse
Affiliation(s)
- Carla G Silva
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Elise Peyre
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Mohit H Adhikari
- Center for Brain and Cognition, Department of Information and Technology, Universitat Pompeu Fabra, Calle Ramon Trias Fargas 25-27, Barcelona 08005, Spain
| | - Sylvia Tielens
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Sebastian Tanco
- VIB-UGent Center for Medical Biotechnologie, VIB, 9000 Ghent, Belgium; Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Petra Van Damme
- VIB-UGent Center for Medical Biotechnologie, VIB, 9000 Ghent, Belgium; Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Lorenza Magno
- Wolfson Institute for Biomedical Research and Department of Cell and Developmental Biology, University College London, London, UK
| | - Nathalie Krusy
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Gulistan Agirman
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Maria M Magiera
- Institut Curie, CNRS UMR3348, PSL Research University, Centre Universitaire, 91400 Orsay, France
| | - Nicoletta Kessaris
- Wolfson Institute for Biomedical Research and Department of Cell and Developmental Biology, University College London, London, UK
| | - Brigitte Malgrange
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Annie Andrieux
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Inserm, U1216, 38000 Grenoble, France
| | - Carsten Janke
- Institut Curie, CNRS UMR3348, PSL Research University, Centre Universitaire, 91400 Orsay, France
| | - Laurent Nguyen
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium.
| |
Collapse
|
89
|
Bulinski JC. Microtubules and Neurodegeneration: The Tubulin Code Sets the Rules of the Road. Curr Biol 2019; 29:R28-R30. [PMID: 30620913 DOI: 10.1016/j.cub.2018.11.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Two recent papers demonstrate that the 'tubulin code' - the pattern of chemical modifications of tubulin along a microtubule - is disrupted upon deletion or mutation of an enzyme, called CCP1, that removes one of these modifications. Ablation of CCP1 interferes with mitochondrial transport and causes human neurodegenerative disease, which may be amenable to pharmacological therapies.
Collapse
Affiliation(s)
- J Chloë Bulinski
- Department of Biological Sciences and Pathology & Cell Biology, Columbia University, 1212 Amsterdam Avenue, New York, NY 10027-2450, USA; Station Biologique de Roscoff, CNRS/Sorbonne Université, Place Georges Teissier, 29688 Roscoff, France.
| |
Collapse
|
90
|
Garapati PV, Zhang J, Rey AJ, Marygold SJ. Towards comprehensive annotation of Drosophila melanogaster enzymes in FlyBase. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2019; 2019:5298334. [PMID: 30689844 PMCID: PMC6343044 DOI: 10.1093/database/bay144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/18/2018] [Indexed: 11/13/2022]
Abstract
The catalytic activities of enzymes can be described using Gene Ontology (GO) terms and Enzyme Commission (EC) numbers. These annotations are available from numerous biological databases and are routinely accessed by researchers and bioinformaticians to direct their work. However, enzyme data may not be congruent between different resources, while the origin, quality and genomic coverage of these data within any one resource are often unclear. GO/EC annotations are assigned either manually by expert curators or inferred computationally, and there is potential for errors in both types of annotation. If such errors remain unchecked, false positive annotations may be propagated across multiple resources, significantly degrading the quality and usefulness of these data. Similarly, the absence of annotations (false negatives) from any one resource can lead to incorrect inferences or conclusions. We are systematically reviewing and enhancing the functional annotation of the enzymes of Drosophila melanogaster, focusing on improvements within the FlyBase (www.flybase.org) database. We have reviewed four major enzyme groups to date: oxidoreductases, lyases, isomerases and ligases. Herein, we describe our review workflow, the improvement in the quality and coverage of enzyme annotations within FlyBase and the wider impact of our work on other related databases.
Collapse
Affiliation(s)
- Phani V Garapati
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Jingyao Zhang
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Alix J Rey
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Steven J Marygold
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| |
Collapse
|
91
|
van der Laan S, Dubra G, Rogowski K. Tubulin glutamylation: a skeleton key for neurodegenerative diseases. Neural Regen Res 2019; 14:1899-1900. [PMID: 31290441 PMCID: PMC6676872 DOI: 10.4103/1673-5374.259611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Siem van der Laan
- Institute of Human Genetics (IGH), Tubulin Code Team, Centre National de la Recherche Scientifique (CNRS)-University of Montpellier, Montpellier, France
| | - Geronimo Dubra
- Institute of Human Genetics (IGH), Tubulin Code Team, Centre National de la Recherche Scientifique (CNRS)-University of Montpellier, Montpellier, France
| | - Krzysztof Rogowski
- Institute of Human Genetics (IGH), Tubulin Code Team, Centre National de la Recherche Scientifique (CNRS)-University of Montpellier, Montpellier, France
| |
Collapse
|
92
|
Giordano T, Gadadhar S, Bodakuntla S, Straub J, Leboucher S, Martinez G, Chemlali W, Bosc C, Andrieux A, Bieche I, Arnoult C, Geimer S, Janke C. Loss of the deglutamylase CCP5 perturbs multiple steps of spermatogenesis and leads to male infertility. J Cell Sci 2019; 132:jcs.226951. [DOI: 10.1242/jcs.226951] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/21/2018] [Indexed: 12/23/2022] Open
Abstract
Sperm cells are highly specialized mammalian cells, and their biogenesis requires unique intracellular structures. Perturbations of spermatogenesis often lead to male infertility. Here we assess the role of a posttranslational modification of tubulin, glutamylation, in spermatogenesis. We show that mice lacking the tubulin deglutamylase CCP5 do not form functional sperm. Spermatids accumulate polyglutamylated tubulin, accompanied by the occurrence of disorganized microtubule arrays, in particular the sperm manchette, fail to re-arrange their intracellular space and accumulate organelles and cytosol, while nuclei condense normally. Strikingly, spermatids lacking CCP5 show supernumerary centrioles, suggesting that glutamylation could control centriole duplication. We show that most of these observed defects are also present in mice in which CCP5 is deleted only in the male germ line, strongly suggesting that they are germ-cell-autonomous. Our findings reveal that polyglutamylation is, beyond its known importance for sperm flagella, and essential regulator of several microtubule-based functions during spermatogenesis. This makes enzymes involved in glutamylation prime candidates for genes involved in male sterility.
Collapse
Affiliation(s)
- Tiziana Giordano
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Sudarshan Gadadhar
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Jonas Straub
- Cell Biology and Electron Microscopy, University of Bayreuth, 95440 Bayreuth, Germany
| | - Sophie Leboucher
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Guillaume Martinez
- Université Grenoble Alpes, Grenoble, F-38000, France
- Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France
| | - Walid Chemlali
- Institut Curie, PSL Research University, Department of Genetics, F-75005, Paris, France
| | - Christophe Bosc
- Université Grenoble Alpes, Grenoble, F-38000, France
- Inserm U1216, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Annie Andrieux
- Université Grenoble Alpes, Grenoble, F-38000, France
- Inserm U1216, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Ivan Bieche
- Institut Curie, PSL Research University, Department of Genetics, F-75005, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, F-75005, Paris, France
| | - Christophe Arnoult
- Université Grenoble Alpes, Grenoble, F-38000, France
- Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France
| | - Stefan Geimer
- Cell Biology and Electron Microscopy, University of Bayreuth, 95440 Bayreuth, Germany
| | - 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
| |
Collapse
|
93
|
Ferreira LT, Figueiredo AC, Orr B, Lopes D, Maiato H. Dissecting the role of the tubulin code in mitosis. Methods Cell Biol 2018; 144:33-74. [PMID: 29804676 DOI: 10.1016/bs.mcb.2018.03.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitosis is an essential process that takes place in all eukaryotes and involves the equal division of genetic material from a parental cell into two identical daughter cells. During mitosis, chromosome movement and segregation are orchestrated by a specialized structure known as the mitotic spindle, composed of a bipolar array of microtubules. The fundamental structure of microtubules comprises of α/β-tubulin heterodimers that associate head-to-tail and laterally to form hollow filaments. In vivo, microtubules are modified by abundant and evolutionarily conserved tubulin posttranslational modifications (PTMs), giving these filaments the potential for a wide chemical diversity. In recent years, the concept of a "tubulin code" has emerged as an extralayer of regulation governing microtubule function. A range of tubulin isoforms, each with a diverse set of PTMs, provides a readable code for microtubule motors and other microtubule-associated proteins. This chapter focuses on the complexity of tubulin PTMs with an emphasis on detyrosination and summarizes the methods currently used in our laboratory to experimentally manipulate these modifications and study their impact in mitosis.
Collapse
Affiliation(s)
- Luísa T Ferreira
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana C Figueiredo
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Bernardo Orr
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Danilo Lopes
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Helder Maiato
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Faculdade de Medicina, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
94
|
Magiera MM, Bodakuntla S, Žiak J, Lacomme S, Marques Sousa P, Leboucher S, Hausrat TJ, Bosc C, Andrieux A, Kneussel M, Landry M, Calas A, Balastik M, Janke C. Excessive tubulin polyglutamylation causes neurodegeneration and perturbs neuronal transport. EMBO J 2018; 37:e100440. [PMID: 30420556 PMCID: PMC6276888 DOI: 10.15252/embj.2018100440] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022] Open
Abstract
Posttranslational modifications of tubulin are emerging regulators of microtubule functions. We have shown earlier that upregulated polyglutamylation is linked to rapid degeneration of Purkinje cells in mice with a mutation in the deglutamylating enzyme CCP1. How polyglutamylation leads to degeneration, whether it affects multiple neuron types, or which physiological processes it regulates in healthy neurons has remained unknown. Here, we demonstrate that excessive polyglutamylation induces neurodegeneration in a cell-autonomous manner and can occur in many parts of the central nervous system. Degeneration of selected neurons in CCP1-deficient mice can be fully rescued by simultaneous knockout of the counteracting polyglutamylase TTLL1. Excessive polyglutamylation reduces the efficiency of neuronal transport in cultured hippocampal neurons, suggesting that impaired cargo transport plays an important role in the observed degenerative phenotypes. We thus establish polyglutamylation as a cell-autonomous mechanism for neurodegeneration that might be therapeutically accessible through manipulation of the enzymes that control this posttranslational modification.
Collapse
Affiliation(s)
- Maria M Magiera
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Satish Bodakuntla
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Jakub Žiak
- Department of Molecular Neurobiology, Institute of Physiology, Czech Academy of Sciences, Prague 4, Czech Republic
- Faculty of Science, Charles University, Prague 2, Czech Republic
| | - Sabrina Lacomme
- Bordeaux Imaging Center, BIC, UMS 3420, Université Bordeaux, Bordeaux, France
| | - Patricia Marques Sousa
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Sophie Leboucher
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Torben J Hausrat
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christophe Bosc
- Grenoble Institut des Neurosciences, GIN, Université Grenoble Alpes, Grenoble, France
- Inserm U1216, Grenoble, France
| | - Annie Andrieux
- Grenoble Institut des Neurosciences, GIN, Université Grenoble Alpes, Grenoble, France
- Inserm U1216, Grenoble, France
| | - Matthias Kneussel
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Landry
- Interdisciplinary Institute for Neuroscience, CNRS UMR5297, Université Bordeaux, Bordeaux, France
| | - André Calas
- Interdisciplinary Institute for Neuroscience, CNRS UMR5297, Université Bordeaux, Bordeaux, France
| | - Martin Balastik
- Department of Molecular Neurobiology, Institute of Physiology, Czech Academy of Sciences, Prague 4, Czech Republic
| | - Carsten Janke
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| |
Collapse
|
95
|
Urbanska P, Joachimiak E, Bazan R, Fu G, Poprzeczko M, Fabczak H, Nicastro D, Wloga D. Ciliary proteins Fap43 and Fap44 interact with each other and are essential for proper cilia and flagella beating. Cell Mol Life Sci 2018; 75:4479-4493. [PMID: 29687140 PMCID: PMC6208767 DOI: 10.1007/s00018-018-2819-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 11/08/2022]
Abstract
Cilia beating is powered by the inner and outer dynein arms (IDAs and ODAs). These multi-subunit macrocomplexes are arranged in two rows on each outer doublet along the entire cilium length, except its distal end. To generate cilia beating, the activity of ODAs and IDAs must be strictly regulated locally by interactions with the dynein arm-associated structures within each ciliary unit and coordinated globally in time and space between doublets and along the axoneme. Here, we provide evidence of a novel ciliary complex composed of two conserved WD-repeat proteins, Fap43p and Fap44p. This complex is adjacent to another WD-repeat protein, Fap57p, and most likely the two-headed inner dynein arm, IDA I1. Loss of either protein results in altered waveform, beat stroke and reduced swimming speed. The ciliary localization of Fap43p and Fap44p is interdependent in the ciliate Tetrahymena thermophila.
Collapse
Affiliation(s)
- Paulina Urbanska
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology PAS, Pasteur 3, 02-093, Warsaw, Poland
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology PAS, Pasteur 3, 02-093, Warsaw, Poland
| | - Rafał Bazan
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology PAS, Pasteur 3, 02-093, Warsaw, Poland
| | - Gang Fu
- Departments of Cell Biology and Biophysics, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, USA
| | - Martyna Poprzeczko
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology PAS, Pasteur 3, 02-093, Warsaw, Poland
| | - Hanna Fabczak
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology PAS, Pasteur 3, 02-093, Warsaw, Poland
| | - Daniela Nicastro
- Departments of Cell Biology and Biophysics, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, USA
| | - Dorota Wloga
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology PAS, Pasteur 3, 02-093, Warsaw, Poland.
| |
Collapse
|
96
|
Bayrak T, Jagtap NS, Erbe A. Review of the Electrical Characterization of Metallic Nanowires on DNA Templates. Int J Mol Sci 2018; 19:E3019. [PMID: 30282940 PMCID: PMC6213931 DOI: 10.3390/ijms19103019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/24/2018] [Accepted: 09/26/2018] [Indexed: 01/29/2023] Open
Abstract
The use of self-assembly techniques may open new possibilities in scaling down electronic circuits to their ultimate limits. Deoxyribonucleic acid (DNA) nanotechnology has already demonstrated that it can provide valuable tools for the creation of nanostructures of arbitrary shape, therefore presenting an ideal platform for the development of nanoelectronic circuits. So far, however, the electronic properties of DNA nanostructures are mostly insulating, thus limiting the use of the nanostructures in electronic circuits. Therefore, methods have been investigated that use the DNA nanostructures as templates for the deposition of electrically conducting materials along the DNA strands. The most simple such structure is given by metallic nanowires formed by deposition of metals along the DNA nanostructures. Here, we review the fabrication and the characterization of the electronic properties of nanowires, which were created using these methods.
Collapse
Affiliation(s)
- Türkan Bayrak
- Institute of Ion Beam Physics and Materials Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Cluster of Excellence Center for Advancing Electronics Dresden (cfaed), TU Dresden, 01062 Dresden, Germany.
| | - Nagesh S Jagtap
- Institute of Ion Beam Physics and Materials Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Artur Erbe
- Institute of Ion Beam Physics and Materials Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Cluster of Excellence Center for Advancing Electronics Dresden (cfaed), TU Dresden, 01062 Dresden, Germany.
| |
Collapse
|
97
|
Bertiaux E, Mallet A, Fort C, Blisnick T, Bonnefoy S, Jung J, Lemos M, Marco S, Vaughan S, Trépout S, Tinevez JY, Bastin P. Bidirectional intraflagellar transport is restricted to two sets of microtubule doublets in the trypanosome flagellum. J Cell Biol 2018; 217:4284-4297. [PMID: 30275108 PMCID: PMC6279389 DOI: 10.1083/jcb.201805030] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/06/2018] [Accepted: 09/21/2018] [Indexed: 12/22/2022] Open
Abstract
Intraflagellar transport (IFT) is the movement of large protein complexes responsible for the construction of cilia and flagella. Using a combination of three-dimensional electron microscopy and high-resolution live imaging, Bertiaux et al. show that IFT takes place on only four microtubule doublets out of the nine available in the trypanosome flagellum. Intraflagellar transport (IFT) is the rapid bidirectional movement of large protein complexes driven by kinesin and dynein motors along microtubule doublets of cilia and flagella. In this study, we used a combination of high-resolution electron and light microscopy to investigate how and where these IFT trains move within the flagellum of the protist Trypanosoma brucei. Focused ion beam scanning electron microscopy (FIB-SEM) analysis of trypanosomes showed that trains are found almost exclusively along two sets of doublets (3–4 and 7–8) and distribute in two categories according to their length. High-resolution live imaging of cells expressing mNeonGreen::IFT81 or GFP::IFT52 revealed for the first time IFT trafficking on two parallel lines within the flagellum. Anterograde and retrograde IFT occurs on each of these lines. At the distal end, a large individual anterograde IFT train is converted in several smaller retrograde trains in the space of 3–4 s while remaining on the same side of the axoneme.
Collapse
Affiliation(s)
- Eloïse Bertiaux
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, Paris, France.,Université Pierre et Marie Curie Paris 6, Cellule Pasteur, Paris, France
| | - Adeline Mallet
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, Paris, France.,Université Pierre et Marie Curie Paris 6, Cellule Pasteur, Paris, France.,UtechS Ultrastructural Bioimaging (Ultrapole), Institut Pasteur, Paris, France
| | - Cécile Fort
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, Paris, France.,Université Pierre et Marie Curie Paris 6, Cellule Pasteur, Paris, France
| | - Thierry Blisnick
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, Paris, France
| | - Serge Bonnefoy
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, Paris, France
| | - Jamin Jung
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, Paris, France
| | - Moara Lemos
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, Paris, France
| | - Sergio Marco
- Université Paris Sud, Université Paris-Saclay, Centre National de la Recherche Scientifique, UMR 9187, Orsay, France.,Institut Curie, Paris Sciences et Lettres Research University, INSERM U1196, Orsay, France
| | - Sue Vaughan
- Department of Biological and Medical Sciences, Faculty of Health and Life Science, Oxford Brookes University, Oxford, UK
| | - Sylvain Trépout
- Université Paris Sud, Université Paris-Saclay, Centre National de la Recherche Scientifique, UMR 9187, Orsay, France.,Institut Curie, Paris Sciences et Lettres Research University, INSERM U1196, Orsay, France
| | - Jean-Yves Tinevez
- UtechS Photonic Bioimaging (Imagopole), Institut Pasteur, Paris, France.,Image Analysis Hub, Institut Pasteur, Paris, France
| | - Philippe Bastin
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, Paris, France
| |
Collapse
|
98
|
He K, Ma X, Xu T, Li Y, Hodge A, Zhang Q, Torline J, Huang Y, Zhao J, Ling K, Hu J. Axoneme polyglutamylation regulated by Joubert syndrome protein ARL13B controls ciliary targeting of signaling molecules. Nat Commun 2018; 9:3310. [PMID: 30120249 PMCID: PMC6098020 DOI: 10.1038/s41467-018-05867-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 07/02/2018] [Indexed: 12/12/2022] Open
Abstract
Tubulin polyglutamylation is a predominant axonemal post-translational modification. However, if and how axoneme polyglutamylation is essential for primary cilia and contribute to ciliopathies are unknown. Here, we report that Joubert syndrome protein ARL13B controls axoneme polyglutamylation, which is marginally required for cilia stability but essential for cilia signaling. ARL13B interacts with RAB11 effector FIP5 to promote cilia import of glutamylase TTLL5 and TTLL6. Hypoglutamylation caused by a deficient ARL13B-RAB11-FIP5 trafficking pathway shows no effect on ciliogenesis, but promotes cilia disassembly and, importantly, impairs cilia signaling by disrupting the proper anchoring of sensory receptors and trafficking of signaling molecules. Remarkably, depletion of deglutamylase CCP5, the predominant cilia deglutamylase, effectively restores hypoglutamylation-induced cilia defects. Our study reveals a paradigm that tubulin polyglutamylation is a major contributor for cilia signaling and suggests a potential therapeutic strategy by targeting polyglutamylation machinery to promote ciliary targeting of signaling machineries and correct signaling defects in ciliopathies.
Collapse
Affiliation(s)
- Kai He
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, 55905, USA
- Mayo Translational PKD Center, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xiaoyu Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, 55905, USA
- Mayo Translational PKD Center, Mayo Clinic, Rochester, MN, 55905, USA
| | - Tao Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, 55905, USA
- Mayo Translational PKD Center, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yan Li
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, 55905, USA
- Mayo Translational PKD Center, Mayo Clinic, Rochester, MN, 55905, USA
| | - Allen Hodge
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Qing Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Julia Torline
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yan Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jian Zhao
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA.
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, 55905, USA.
- Mayo Translational PKD Center, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
99
|
Tang NH, Jin Y. Shaping neurodevelopment: distinct contributions of cytoskeletal proteins. Curr Opin Neurobiol 2018; 51:111-118. [PMID: 29574219 PMCID: PMC6066413 DOI: 10.1016/j.conb.2018.02.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/20/2018] [Accepted: 02/27/2018] [Indexed: 12/28/2022]
Abstract
Development of a neuron critically depends on the organization of its cytoskeleton. Cytoskeletal components, such as tubulins and actins, have the remarkable ability to organize themselves into filaments and networks to support specialized and compartmentalized functions. Alterations in cytoskeletal proteins have long been associated with a variety of neurodevelopmental disorders. This review focuses on recent findings, primarily from forward genetic screens in Caenorhabditis elegans that illustrate how different tubulin protein isotypes can play distinct roles in neuronal development and function. Additionally, we discuss studies revealing new regulators of the actin cytoskeleton, and highlight recent technological advances in in vivo imaging and functional dissection of the neuronal cytoskeleton.
Collapse
Affiliation(s)
- Ngang Heok Tang
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| | - Yishi Jin
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
100
|
Luo WW, Shu HB. Delicate regulation of the cGAS-MITA-mediated innate immune response. Cell Mol Immunol 2018; 15:666-675. [PMID: 29456253 PMCID: PMC6123429 DOI: 10.1038/cmi.2016.51] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 12/14/2022] Open
Abstract
Although it has long been demonstrated that cytosolic DNA is a potent immune stimulant, it is only in recent years that the molecular mechanisms of DNA-stimulated innate immune responses have emerged. Studies have established critical roles for the DNA sensor cyclic GMP-AMP synthase (cGAS) and the adapter protein MITA/STING in the innate immune response to cytosolic DNA or DNA viruses. Although the regulation of cGAS-MITA/STING-mediated signaling remains to be fully investigated, understanding the processes involved may help to explain the mechanisms of innate immune signaling events and perhaps autoinflammatory diseases and to provide potential therapeutic targets for drug intervention. In this review, we summarize recent progress on the regulation of the cGAS-MITA/STING-mediated innate immune response to DNA viruses at the organelle-trafficking, post-translational and transcriptional levels.
Collapse
Affiliation(s)
- Wei-Wei Luo
- Medical Research Institute, Collaborative Innovation Center for Viral Immunology, Wuhan University, Wuhan, 430071, China
| | - Hong-Bing Shu
- Medical Research Institute, Collaborative Innovation Center for Viral Immunology, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|