251
|
Kelliher MT, Saunders HA, Wildonger J. Microtubule control of functional architecture in neurons. Curr Opin Neurobiol 2019; 57:39-45. [PMID: 30738328 DOI: 10.1016/j.conb.2019.01.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 01/20/2023]
Abstract
Neurons are exquisitely polarized cells whose structure and function relies on microtubules. Microtubules in signal-receiving dendrites and signal-sending axons differ in their organization and microtubule-associated proteins. These differences, coupled with microtubule post-translational modifications, combine to locally regulate intracellular transport, morphology, and function. Recent discoveries provide new insight into the regulation of non-centrosomal microtubule arrays in neurons, the relationship between microtubule acetylation and mechanosensation, and the spatial patterning of microtubules that regulates motor activity and cargo delivery in axons and dendrites. Together, these new studies bring us closer to understanding how microtubule function is locally tuned to match the specialized tasks associated with signal reception and transmission.
Collapse
Affiliation(s)
- Michael T Kelliher
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Harriet Aj Saunders
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jill Wildonger
- Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
252
|
Yang L, Zhao L, Cui L, Huang Y, Ye J, Zhang Q, Jiang X, Zhang D, Huang Y. Decreased α-tubulin acetylation induced by an acidic environment impairs autophagosome formation and leads to rat cardiomyocyte injury. J Mol Cell Cardiol 2019; 127:143-153. [DOI: 10.1016/j.yjmcc.2018.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 12/02/2018] [Accepted: 12/20/2018] [Indexed: 11/26/2022]
|
253
|
Zeng QZ, Yang F, Li CG, Xu LH, He XH, Mai FY, Zeng CY, Zhang CC, Zha QB, Ouyang DY. Paclitaxel Enhances the Innate Immunity by Promoting NLRP3 Inflammasome Activation in Macrophages. Front Immunol 2019; 10:72. [PMID: 30761140 PMCID: PMC6361797 DOI: 10.3389/fimmu.2019.00072] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/11/2019] [Indexed: 12/22/2022] Open
Abstract
Microtubules play critical roles in regulating the activation of NLRP3 inflammasome and microtubule-destabilizing agents such as colchicine have been shown to suppress the activation of this inflammasome. However, it remains largely unknown whether paclitaxel, a microtubule-stabilizing agent being used in cancer therapy, has any influences on NLRP3 inflammasome activation. Here we showed that paclitaxel pre-treatment greatly enhanced ATP- or nigericin-induced NLRP3 inflammasome activation as indicated by increased release of cleaved caspase-1 and mature IL-1β, enhanced formation of ASC speck, and increased gasdermin D cleavage and pyroptosis. Paclitaxel time- and dose-dependently induced α-tubulin acetylation in LPS-primed murine and human macrophages and further increased ATP- or nigericin-induced α-tubulin acetylation. Such increased α-tubulin acetylation was significantly suppressed either by resveratrol or NAD+ (coenzyme required for deacetylase activity of SIRT2), or by genetic knockdown of MEC-17 (gene encoding α-tubulin acetyltransferase 1). Concurrently, the paclitaxel-mediated enhancement of NLRP3 inflammasome activation was significantly suppressed by resveratrol, NAD+, or MEC-17 knockdown, indicating the involvement of paclitaxel-induced α-tubulin acetylation in the augmentation of NLRP3 inflammasome activation. Similar to paclitaxel, epothilone B that is another microtubule-stabilizing agent also induced α-tubulin acetylation and increased NLRP3 inflammasome activation in macrophages in response to ATP treatment. Consistent with the in vitro results, intraperitoneal administration of paclitaxel significantly increased serum IL-1β levels, reduced bacterial burden, dampened infiltration of inflammatory cells in the liver, and improved animal survival in a mouse model of bacterial infection. Collectively, our data indicate that paclitaxel potentiated NLRP3 inflammasome activation by inducing α-tubulin acetylation and thereby conferred enhanced antibacterial innate responses, suggesting its potential application against pathogenic infections beyond its use as a chemotherapeutic agent.
Collapse
Affiliation(s)
- Qiong-Zhen Zeng
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Fan Yang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Chen-Guang Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Li-Hui Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xian-Hui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Feng-Yi Mai
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Chen-Ying Zeng
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Cheng-Cheng Zhang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qing-Bing Zha
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dong-Yun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
254
|
Ying G, Frederick JM, Baehr W. Deletion of both centrin 2 (CETN2) and CETN3 destabilizes the distal connecting cilium of mouse photoreceptors. J Biol Chem 2019; 294:3957-3973. [PMID: 30647131 DOI: 10.1074/jbc.ra118.006371] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/09/2019] [Indexed: 02/03/2023] Open
Abstract
Centrins (CETN1-4) are ubiquitous and conserved EF-hand-family Ca2+-binding proteins associated with the centrosome, basal body, and transition zone. Deletion of CETN1 or CETN2 in mice causes male infertility or dysosmia, respectively, without affecting photoreceptor function. However, it remains unclear to what extent centrins are redundant with each other in photoreceptors. Here, to explore centrin redundancy, we generated Cetn3 GT/GT single-knockout and Cetn2 -/-;Cetn3 GT/GT double-knockout mice. Whereas the Cetn3 deletion alone did not affect photoreceptor function, simultaneous ablation of Cetn2 and Cetn3 resulted in attenuated scotopic and photopic electroretinography (ERG) responses in mice at 3 months of age, with nearly complete retina degeneration at 1 year. Removal of CETN2 and CETN3 activity from the lumen of the connecting cilium (CC) destabilized the photoreceptor axoneme and reduced the CC length as early as postnatal day 22 (P22). In Cetn2 -/-;Cetn3 GT/GT double-knockout mice, spermatogenesis-associated 7 (SPATA7), a key organizer of the photoreceptor-specific distal CC, was depleted gradually, and CETN1 was condensed to the mid-segment of the CC. Ultrastructural analysis revealed that in this double knockout, the axoneme of the CC expanded radially at the distal end, with vertically misaligned outer segment discs and membrane whorls. These observations suggest that CETN2 and CETN3 cooperate in stabilizing the CC/axoneme structure.
Collapse
Affiliation(s)
- Guoxin Ying
- From the Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, Utah 84132,
| | - Jeanne M Frederick
- From the Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, Utah 84132
| | - Wolfgang Baehr
- From the Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, Utah 84132, .,the Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah 84112, and.,the Department of Biology, University of Utah, Salt Lake City, Utah 84132
| |
Collapse
|
255
|
Sadoul K, Joubert C, Michallet S, Nolte E, Peronne L, Ramirez-Rios S, Ribba AS, Lafanechère L. [On the road to deciphering the tubulin code: focus on acetylation and detyrosination]. Med Sci (Paris) 2019; 34:1047-1055. [PMID: 30623774 DOI: 10.1051/medsci/2018295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Microtubules are cytoskeletal fibers formed by the assembly of α- and β-tubulin heterodimers. They contribute to cell morphology, mobility and polarity, as well as to cellular transport processes and cell division. The microtubular network constantly adapts to cellular needs and may be composed of very dynamic or more stable microtubules. To regulate their diverse functions in a spatio-temporal manner, microtubules are subjected to numerous reversible post-translational modifications, which generate the "tubulin code". This review focuses on two modifications characteristic of stable microtubules - acetylation and detyrosination of α-tubulin - and their deregulation in certain pathologies.
Collapse
Affiliation(s)
- Karin Sadoul
- Régulation et pharmacologie du cytosquelette, Institut pour l'avancée des biosciences, Université Grenoble Alpes ; Inserm U 1209 ; CNRS 5309, Allée des Alpes, 38700 La Tronche, France
| | - Clotilde Joubert
- Régulation et pharmacologie du cytosquelette, Institut pour l'avancée des biosciences, Université Grenoble Alpes ; Inserm U 1209 ; CNRS 5309, Allée des Alpes, 38700 La Tronche, France
| | - Sophie Michallet
- Régulation et pharmacologie du cytosquelette, Institut pour l'avancée des biosciences, Université Grenoble Alpes ; Inserm U 1209 ; CNRS 5309, Allée des Alpes, 38700 La Tronche, France
| | - Elsie Nolte
- Régulation et pharmacologie du cytosquelette, Institut pour l'avancée des biosciences, Université Grenoble Alpes ; Inserm U 1209 ; CNRS 5309, Allée des Alpes, 38700 La Tronche, France
| | - Lauralie Peronne
- Régulation et pharmacologie du cytosquelette, Institut pour l'avancée des biosciences, Université Grenoble Alpes ; Inserm U 1209 ; CNRS 5309, Allée des Alpes, 38700 La Tronche, France
| | - Sacnicté Ramirez-Rios
- Régulation et pharmacologie du cytosquelette, Institut pour l'avancée des biosciences, Université Grenoble Alpes ; Inserm U 1209 ; CNRS 5309, Allée des Alpes, 38700 La Tronche, France
| | - Anne-Sophie Ribba
- Régulation et pharmacologie du cytosquelette, Institut pour l'avancée des biosciences, Université Grenoble Alpes ; Inserm U 1209 ; CNRS 5309, Allée des Alpes, 38700 La Tronche, France
| | - Laurence Lafanechère
- Régulation et pharmacologie du cytosquelette, Institut pour l'avancée des biosciences, Université Grenoble Alpes ; Inserm U 1209 ; CNRS 5309, Allée des Alpes, 38700 La Tronche, France
| |
Collapse
|
256
|
Guo T, Dakkak D, Rodriguez-Martin T, Noble W, Hanger DP. A pathogenic tau fragment compromises microtubules, disrupts insulin signaling and induces the unfolded protein response. Acta Neuropathol Commun 2019; 7:2. [PMID: 30606258 PMCID: PMC6318896 DOI: 10.1186/s40478-018-0651-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 12/13/2018] [Indexed: 12/02/2022] Open
Abstract
Human tauopathies including Alzheimer’s disease, progressive supranuclear palsy and related disorders, are characterized by deposition of pathological forms of tau, synaptic dysfunction and neuronal loss. We have previously identified a pathogenic C-terminal tau fragment (Tau35) that is associated with human tauopathy. However, it is not known how tau fragmentation affects critical molecular processes in cells and contributes to impaired physiological function. Chinese hamster ovary (CHO) cells and new CHO cell lines stably expressing Tau35 or full-length human tau were used to compare the effects of disease-associated tau cleavage on tau function and signaling pathways. Western blots, microtubule-binding assays and immunofluorescence labeling were used to examine the effects of Tau35 on tau function and on signaling pathways in CHO cells. We show that Tau35 undergoes aberrant phosphorylation when expressed in cells. Although Tau35 contain the entire microtubule-binding region, the lack of the amino terminal half of tau results in a marked reduction in microtubule binding and defective microtubule organization in cells. Notably, Tau35 attenuates insulin-mediated activation of Akt and a selective inhibitory phosphorylation of glycogen synthase kinase-3. Moreover, Tau35 activates ribosomal protein S6 kinase beta-1 signaling and the unfolded protein response, leading to insulin resistance in cells. Tau35 has deleterious effects on signaling pathways that mediate pathological changes and insulin resistance, suggesting a mechanism through which N-terminal cleavage of tau leads to the development and progression of tau pathology in human tauopathy. Our findings highlight the importance of the N-terminal region of tau for its normal physiological function. Furthermore, we show that pathogenic tau cleavage induces tau phosphorylation, resulting in impaired microtubule binding, disruption of insulin signaling and activation of the unfolded protein response. Since insulin resistance is a feature of several tauopathies, this work suggests new potential targets for therapeutic intervention.
Collapse
|
257
|
Dilan TL, Singh RK, Saravanan T, Moye A, Goldberg AFX, Stoilov P, Ramamurthy V. Bardet-Biedl syndrome-8 (BBS8) protein is crucial for the development of outer segments in photoreceptor neurons. Hum Mol Genet 2019; 27:283-294. [PMID: 29126234 DOI: 10.1093/hmg/ddx399] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/06/2017] [Indexed: 11/15/2022] Open
Abstract
Bardet-Biedl syndrome (BBS) is an autosomal recessive ciliopathy characterized by developmental abnormalities and vision loss. To date, mutations in 21 genes have been linked to BBS. The products of eight of these BBS genes form a stable octameric complex termed the BBSome. Mutations in BBS8, a component of the BBSome, cause early vision loss, but the role of BBS8 in supporting vision is not known. To understand the mechanisms by which BBS8 supports rod and cone photoreceptor function, we generated animal models lacking BBS8. The loss of BBS8 protein led to concomitant decrease in the levels of BBSome subunits, BBS2 and BBS5 and increase in the levels of the BBS1 and BBS4 subunits. BBS8 ablation was associated with severe reduction of rod and cone photoreceptor function and progressive degeneration of each photoreceptor subtype. We observed disorganized and shortened photoreceptor outer segments (OS) at post-natal day 10 as the OS elaborates. Interestingly, loss of BBS8 led to changes in the distribution of photoreceptor axonemal proteins and hyper-acetylation of ciliary microtubules. In contrast to properly localized phototransduction machinery, we observed OS accumulation of syntaxin3, a protein normally found in the cytoplasm and the synaptic termini. In conclusion, our studies demonstrate the requirement for BBS8 in early development and elaboration of ciliated photoreceptor OS, explaining the need for BBS8 in normal vision. The findings from our study also imply that early targeting of both rods and cones in BBS8 patients is crucial for successful restoration of vision.
Collapse
Affiliation(s)
- Tanya L Dilan
- Departments of Ophthalmology, West Virginia University, Morgantown, WA 26506, USA.,Biochemistry, West Virginia University, Morgantown, WA 26506, USA
| | - Ratnesh K Singh
- Departments of Ophthalmology, West Virginia University, Morgantown, WA 26506, USA.,Biochemistry, West Virginia University, Morgantown, WA 26506, USA
| | | | - Abigail Moye
- Departments of Ophthalmology, West Virginia University, Morgantown, WA 26506, USA.,Biochemistry, West Virginia University, Morgantown, WA 26506, USA
| | | | - Peter Stoilov
- Biochemistry, West Virginia University, Morgantown, WA 26506, USA
| | - Visvanathan Ramamurthy
- Departments of Ophthalmology, West Virginia University, Morgantown, WA 26506, USA.,Biochemistry, West Virginia University, Morgantown, WA 26506, USA.,Rockefeller Neurosciences Institute, West Virginia University, Morgantown, WA 26506, USA
| |
Collapse
|
258
|
Zheng J, Liu H, Zhu L, Chen Y, Zhao H, Zhang W, Li F, Xie L, Yan X, Zhu X. Microtubule-bundling protein Spef1 enables mammalian ciliary central apparatus formation. J Mol Cell Biol 2019; 11:67-77. [PMID: 30535028 DOI: 10.1093/jmcb/mjy014] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/22/2018] [Indexed: 01/10/2023] Open
Abstract
Cilia are cellular protrusions containing nine microtubule (MT) doublets and function to propel cell movement or extracellular liquid flow through beating or sense environmental stimuli through signal transductions. Cilia require the central pair (CP) apparatus, consisting of two CP MTs covered with projections of CP proteins, for planar strokes. How the CP MTs of such '9 + 2' cilia are constructed, however, remains unknown. Here we identify Spef1, an evolutionarily conserved microtubule-bundling protein, as a core CP MT regulator in mammalian cilia. Spef1 was selectively expressed in mammalian cells with 9 + 2 cilia and specifically localized along the CP. Its depletion in multiciliated mouse ependymal cells by RNAi completely abolished the CP MTs and markedly attenuated ciliary localizations of CP proteins such as Hydin and Spag6, resulting in rotational beat of the ependymal cilia. Spef1, which binds to MTs through its N-terminal calponin-homologous domain, formed homodimers through its C-terminal coiled coil region to bundle and stabilize MTs. Disruption of either the MT-binding or the dimerization activity abolished the ability of exogenous Spef1 to restore the structure and functions of the CP apparatus. We propose that Spef1 bundles and stabilizes central MTs to enable the assembly and functions of the CP apparatus.
Collapse
Affiliation(s)
- Jianqun Zheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Hao Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Lei Zhu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Yawen Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Huijie Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
| | - Wei Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
| | - Fan Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Lele Xie
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
| | - Xiumin Yan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
| | - Xueliang Zhu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai0, China
- University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
259
|
Singh R, Holz PS, Roth K, Hupfer A, Meissner W, Müller R, Buchholz M, Gress TM, Elsässer HP, Jacob R, Lauth M. DYRK1B regulates Hedgehog-induced microtubule acetylation. Cell Mol Life Sci 2019; 76:193-207. [PMID: 30317528 PMCID: PMC11105311 DOI: 10.1007/s00018-018-2942-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 09/25/2018] [Accepted: 10/08/2018] [Indexed: 01/12/2023]
Abstract
The posttranslational modification (PTM) of tubulin subunits is important for the physiological functions of the microtubule (MT) cytoskeleton. Although major advances have been made in the identification of enzymes carrying out MT-PTMs, little knowledge is available on how intercellular signaling molecules and their associated pathways regulate MT-PTM-dependent processes inside signal-receiving cells. Here we show that Hedgehog (Hh) signaling, a paradigmatic intercellular signaling system, affects the MT acetylation state in mammalian cells. Mechanistically, Hh pathway activity increases the levels of the MT-associated DYRK1B kinase, resulting in the inhibition of GSK3β through phosphorylation of Serine 9 and the subsequent suppression of HDAC6 enzyme activity. Since HDAC6 represents a major tubulin deacetylase, its inhibition increases the levels of acetylated MTs. Through the activation of DYRK1B, Hh signaling facilitates MT-dependent processes such as intracellular mitochondrial transport, mesenchymal cell polarization or directed cell migration. Taken together, we provide evidence that intercellular communication through Hh signals can regulate the MT cytoskeleton and contribute to MT-dependent processes by affecting the level of tubulin acetylation.
Collapse
Affiliation(s)
- Rajeev Singh
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor- and Immune Biology (ZTI), Philipps University, Hans-Meerwein-Str. 3, 35043, Marburg, Germany
| | - Philipp Simon Holz
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor- and Immune Biology (ZTI), Philipps University, Hans-Meerwein-Str. 3, 35043, Marburg, Germany
| | - Katrin Roth
- Imaging Core Facility, Center for Tumor- and Immune Biology (ZTI), Philipps University, Hans-Meerwein-Str. 3, 35043, Marburg, Germany
| | - Anna Hupfer
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor- and Immune Biology (ZTI), Philipps University, Hans-Meerwein-Str. 3, 35043, Marburg, Germany
| | - Wolfgang Meissner
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor- and Immune Biology (ZTI), Philipps University, Hans-Meerwein-Str. 3, 35043, Marburg, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor- and Immune Biology (ZTI), Philipps University, Hans-Meerwein-Str. 3, 35043, Marburg, Germany
| | - Malte Buchholz
- Clinic for Gastroenterology, Endocrinology, Metabolism and Infectiology, Philipps University, Marburg, Germany
| | - Thomas M Gress
- Clinic for Gastroenterology, Endocrinology, Metabolism and Infectiology, Philipps University, Marburg, Germany
| | - Hans-Peter Elsässer
- Institute of Cytobiology and Cytopathology, Philipps University, Robert Koch Str. 6, 35037, Marburg, Germany
| | - Ralf Jacob
- Institute of Cytobiology and Cytopathology, Philipps University, Robert Koch Str. 6, 35037, Marburg, Germany
| | - Matthias Lauth
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor- and Immune Biology (ZTI), Philipps University, Hans-Meerwein-Str. 3, 35043, Marburg, Germany.
| |
Collapse
|
260
|
Donhauser ZJ, Appadoo V, Kliman EJ, Jobs WB, Sheffield EC. Structural Changes in Tubulin Sheets Caused by Immobilization on Solid Supports. ACS OMEGA 2018; 3:18196-18202. [PMID: 30613819 PMCID: PMC6312633 DOI: 10.1021/acsomega.8b02475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/11/2018] [Indexed: 06/09/2023]
Abstract
In the presence of zinc, the protein tubulin assembles into two-dimensional sheets that are a useful model system for the study of both tubulin and microtubule structure. Tubulin sheets present an ideal protein structure for study with atomic force microscopy because they contain a two-dimensional crystalline protein lattice and retain many of the structural features of tubulin and microtubules. However, high-resolution imaging requires nonperturbative immobilization onto an appropriate imaging substrate. In this report, several substrates commonly used for scanning probe microscopy are evaluated for their ability to effectively immobilize tubulin sheets: mica, gold, highly ordered pyrolytic graphite, and carbon-coated electron microscopy grids. We hypothesize that the different intermolecular interactions presented by these substrates will affect the morphology of adsorbed tubulin sheets as well as the amount of other contaminating adsorbates. Tubulin sheets were successfully imaged on all of these substrates and structural characterization is reported. The most consistent results were obtained on carbon-coated electron microscopy grids, which preserved fine structural features of the sheets and had the least amount of contamination from the adsorption of unpolymerized tubulin. Images of tubulin sheets obtained with atomic force microscopy also compare favorably with published electron micrographs of sheets produced using similar procedures. This work demonstrates the importance of assessing substrate effects when studying two-dimensional protein crystals and identifies suitable substrates for immobilizing tubulin sheets.
Collapse
Affiliation(s)
| | | | - Elysa J. Kliman
- Vassar College, 124 Raymond Avenue, Poughkeepsie, New York 12604, United States
| | - William B. Jobs
- Vassar College, 124 Raymond Avenue, Poughkeepsie, New York 12604, United States
| | - Evan C. Sheffield
- Vassar College, 124 Raymond Avenue, Poughkeepsie, New York 12604, United States
| |
Collapse
|
261
|
Baehr W, Hanke-Gogokhia C, Sharif A, Reed M, Dahl T, Frederick JM, Ying G. Insights into photoreceptor ciliogenesis revealed by animal models. Prog Retin Eye Res 2018; 71:26-56. [PMID: 30590118 DOI: 10.1016/j.preteyeres.2018.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 12/10/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022]
Abstract
Photoreceptors are polarized neurons, with very specific subcellular compartmentalization and unique requirements for protein expression and trafficking. Each photoreceptor contains an outer segment, the site of photon capture that initiates vision, an inner segment that houses the biosynthetic machinery and a synaptic terminal for signal transmission to downstream neurons. Outer segments and inner segments are connected by a connecting cilium (CC), the equivalent of a transition zone (TZ) of primary cilia. The connecting cilium is part of the basal body/axoneme backbone that stabilizes the outer segment. This report will update the reader on late developments in photoreceptor ciliogenesis and transition zone formation, specifically in mouse photoreceptors, focusing on early events in photoreceptor ciliogenesis. The connecting cilium, an elongated and narrow structure through which all outer segment proteins and membrane components must traffic, functions as a gate that controls access to the outer segment. Here we will review genes and their protein products essential for basal body maturation and for CC/TZ genesis, sorted by phenotype. Emphasis is given to naturally occurring mouse mutants and gene knockouts that interfere with CC/TZ formation and ciliogenesis.
Collapse
Affiliation(s)
- Wolfgang Baehr
- Department of Ophthalmology and Visual Sciences, University of Utah Health Sciences, Salt Lake City, UT, 84132, USA.
| | - Christin Hanke-Gogokhia
- Department of Ophthalmology and Visual Sciences, University of Utah Health Sciences, Salt Lake City, UT, 84132, USA
| | - Ali Sharif
- Department of Ophthalmology and Visual Sciences, University of Utah Health Sciences, Salt Lake City, UT, 84132, USA
| | - Michelle Reed
- Department of Ophthalmology and Visual Sciences, University of Utah Health Sciences, Salt Lake City, UT, 84132, USA
| | - Tiffanie Dahl
- Department of Ophthalmology and Visual Sciences, University of Utah Health Sciences, Salt Lake City, UT, 84132, USA
| | - Jeanne M Frederick
- Department of Ophthalmology and Visual Sciences, University of Utah Health Sciences, Salt Lake City, UT, 84132, USA
| | - Guoxin Ying
- Department of Ophthalmology and Visual Sciences, University of Utah Health Sciences, Salt Lake City, UT, 84132, USA
| |
Collapse
|
262
|
Pradhan K, Das G, Mondal P, Khan J, Barman S, Ghosh S. Genesis of Neuroprotective Peptoid from Aβ30-34 Inhibits Aβ Aggregation and AChE Activity. ACS Chem Neurosci 2018; 9:2929-2940. [PMID: 30036464 DOI: 10.1021/acschemneuro.8b00071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aβ peptide and hyper-phosphorylated microtubule associated protein (Tau) aggregation causes severe damage to both the neuron membrane and key signal processing microfilament (microtubule) in Alzheimer's disease (AD) brains. To date, the key challenge is to develop nontoxic, proteolytically stable amyloid inhibitors, which can simultaneously target multiple pathways involved in AD. Various attempts have been made in this direction; however, clinical outcomes of those attempts have been reported to be poor. Thus, we choose development of peptoid (N-substituted glycine oligomers)-based leads as potential AD therapeutics, which are easy to synthesize, found to be proteolytically stable, and exhibit excellent bioavailability. In this paper, we have designed and synthesized a new short peptoid for amyloid inhibition from 30-34 hydrophobic pocket of amyloid beta (Aβ) peptide. The peptoid selectively binds with 17-21 hydrophobic region of Aβ and inhibits Aβ fibril formation. Various in vitro assays suggested that our AI peptoid binds with tubulin/microtubule and promotes its polymerization and stability. This peptoid also inhibits AChE-induced Aβ fibril formation and provides significant neuroprotection against toxicity generated by nerve growth factor (NGF) deprived neurons derived from rat adrenal pheochromocytoma (PC12) cell line. Moreover, this peptoid shows serum stability and is noncytotoxic to primary rat cortical neurons.
Collapse
Affiliation(s)
- Krishnangsu Pradhan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Gaurav Das
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Prasenjit Mondal
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Juhee Khan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Surajit Barman
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Surajit Ghosh
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| |
Collapse
|
263
|
Ilan Y. Microtubules: From understanding their dynamics to using them as potential therapeutic targets. J Cell Physiol 2018; 234:7923-7937. [PMID: 30536951 DOI: 10.1002/jcp.27978] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023]
Abstract
Microtubules (MT) and actin microfilaments are dynamic cytoskeleton components involved in a range of intracellular processes. MTs play a role in cell division, beating of cilia and flagella, and intracellular transport. Over the past decades, much knowledge has been gained regarding MT function and structure, and its role in underlying disease progression. This makes MT potential therapeutic targets for various disorders. Disturbances in MT and their associated proteins are the underlying cause of diseases such as Alzheimer's disease, cancer, and several genetic diseases. Some of the advances in the field of MT research, as well as the potenti G beta gamma, is needed al uses of MT-targeting agents in various conditions have been reviewed here.
Collapse
Affiliation(s)
- Yaron Ilan
- Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
264
|
Hanson K, Tian N, Vickers JC, King AE. The HDAC6 Inhibitor Trichostatin A Acetylates Microtubules and Protects Axons From Excitotoxin-Induced Degeneration in a Compartmented Culture Model. Front Neurosci 2018; 12:872. [PMID: 30555293 PMCID: PMC6282003 DOI: 10.3389/fnins.2018.00872] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/08/2018] [Indexed: 12/15/2022] Open
Abstract
Axon degeneration has been implicated as a pathological process in several neurodegenerative diseases and acquired forms of neural injury. We have previously shown that stabilizing microtubules can protect axons against excitotoxin-induced fragmentation, however, the alterations of microtubules following excitotoxicity that results in axon degeneration are currently unknown. Hence, this study investigated whether excitotoxicity affects the post-translational modifications of microtubules and microtubule-associated proteins, and whether reversing these changes has the potential to rescue axons from degeneration. To investigate microtubule alterations, primary mouse cortical neurons at 10 days in vitro were treated with 10 or 25 μM kainic acid to induce excitotoxicity and axon degeneration. Post-translational modifications of microtubules and associated proteins were examined at 6 h following kainic acid exposure, relative to axon degeneration. While there were no changes to tyrosinated tubulin or MAP1B, acetylated tubulin was significantly (p < 0.05) decreased by 40% at 6 h post-treatment. To determine whether increasing microtubule acetylation prior to kainic acid exposure could prevent axon fragmentation, we investigated the effect of reducing microtubule deacetylation with the HDAC6 inhibitor, trichostatin A. We found that trichostatin A prevented kainic acid-induced microtubule deacetylation and significantly (p < 0.05) protected axons from fragmentation. These data suggest that microtubule acetylation is a potential target for axonal protection where excitotoxicity may play a role in neuronal degeneration.
Collapse
Affiliation(s)
- Kelsey Hanson
- Wicking Dementia Research and Education Centre, College of Health, University of Tasmania, Hobart, TAS, Australia
| | - Nan Tian
- Wicking Dementia Research and Education Centre, College of Health, University of Tasmania, Hobart, TAS, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, College of Health, University of Tasmania, Hobart, TAS, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, College of Health, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
265
|
Shiki A, Inoh Y, Yokawa S, Furuno T. Promotion of microtubule acetylation plays an important role in degranulation of antigen-activated mast cells. Inflamm Res 2018; 68:181-184. [DOI: 10.1007/s00011-018-1203-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/08/2018] [Accepted: 11/19/2018] [Indexed: 11/29/2022] Open
|
266
|
Mondal P, Gupta V, Das G, Pradhan K, Khan J, Gharai PK, Ghosh S. Peptide-Based Acetylcholinesterase Inhibitor Crosses the Blood-Brain Barrier and Promotes Neuroprotection. ACS Chem Neurosci 2018; 9:2838-2848. [PMID: 30015476 DOI: 10.1021/acschemneuro.8b00253] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Design and development of acetylcholinesterase (AChE) inhibitor has tremendous implications in the treatment of Alzheimer's disease (AD). Here, we have adopted a computational approach for the design of a peptide based AChE inhibitor from its active site. We identified an octapeptide, which interacts with the catalytic anionic site (CAS) of AChE enzyme and inhibits its activity. Interestingly, this peptide also inhibits amyloid aggregation through its interaction at the 17-21 region of amyloid-beta (Aβ) and stabilizes microtubules by interacting with tubulin as well. Eventually, in the PC12 derived neurons, it shows noncytotoxicity, promotes neurite out-growth, stabilizes intracellular microtubules, and confers significant neuroprotection even upon withdrawal of nerve growth factor (NGF). Further, results reveal that this peptide possesses good serum stability, crosses the blood-brain barrier (BBB), and maintains the healthy architecture of the primary cortical neurons. This work shows discovery of an excellent peptide-based AChE inhibitor with additional potential as a microtubule stabilizer, which will pave the way for the development of potential anti-AD therapeutics in the near future.
Collapse
Affiliation(s)
- Prasenjit Mondal
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Varsha Gupta
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India
| | - Gaurav Das
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Krishnangsu Pradhan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India
| | - Juhee Khan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India
| | - Prabir Kumar Gharai
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India
| | - Surajit Ghosh
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| |
Collapse
|
267
|
Roll-Mecak A. How cells exploit tubulin diversity to build functional cellular microtubule mosaics. Curr Opin Cell Biol 2018; 56:102-108. [PMID: 30466050 DOI: 10.1016/j.ceb.2018.10.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/16/2018] [Accepted: 10/31/2018] [Indexed: 11/26/2022]
Abstract
Cellular microtubules are mosaic polymers assembled from multiple αβ-tubulin isoforms bearing chemically diverse posttranslational modifications. This tubulin diversity constitutes a combinatorial code that regulates microtubule interactions with cellular effectors and alters their intrinsic dynamic and mechanical properties. Cells generate stereotyped and complex tubulin modification patterns that are important for their specialized functions. Here we give a brief overview of the tubulin genetic and chemical diversity and highlight recent advances in our understanding of how the tubulin code regulates essential biological processes ranging from intracellular cargo transport, to cell division and cardiomyocyte contraction. Finally, we speculate on the molecular mechanisms for the generation and maintenance of the complex stereotyped modification patterns that form cellular microtubule mosaics.
Collapse
Affiliation(s)
- Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA; Biophysics Center, National Heart, Lung and Blood Institute, MD 20892, USA.
| |
Collapse
|
268
|
Microtubule lattice plasticity. Curr Opin Cell Biol 2018; 56:88-93. [PMID: 30415187 DOI: 10.1016/j.ceb.2018.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/21/2018] [Indexed: 01/06/2023]
Abstract
In classical microtubule dynamic instability, the dynamics of the built polymer depend only on the nucleotide state of its individual tubulin molecules. Recent work is overturning this view, pointing instead towards lattice plasticity, in which the fine-structure and mechanics of the microtubule lattice are emergent properties that depend not only on the nucleotide state of each tubulin, but also on the nucleotide states of its neighbours, on its and their isotypes, and on interacting proteins, drugs, local mechanical strain, post translational modifications, packing defects and solvent conditions. In lattice plasticity models, the microtubule is an allosteric molecular collective that integrates multiple mechanochemical inputs and responds adaptively by adjusting its conformation, stiffness and dynamics.
Collapse
|
269
|
Gasic I, Mitchison TJ. Autoregulation and repair in microtubule homeostasis. Curr Opin Cell Biol 2018; 56:80-87. [PMID: 30415186 DOI: 10.1016/j.ceb.2018.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/24/2018] [Accepted: 10/21/2018] [Indexed: 10/27/2022]
Abstract
Even in the face of damaging insults, most cells maintain stability over time through multiple homeostatic pathways, including maintenance of the microtubule cytoskeleton that is fundamental to numerous cellular processes. The dynamic instability-perpetual growth and shrinkage-is the best-known microtubule regulatory pathway, which allows rapid rebuilding of the microtubule cytoskeleton in response to internal or external cues. Much less investigated is homeostatic regulation through availability of α-β tubulin heterodimers-microtubules' main building blocks-which influences total mass and dynamic behavior of microtubules. Finally, the most recently discovered is microtubule homeostasis through self-repair, where new GTP-bound tubulin heterodimers replace the lost ones in the microtubule lattice. In this review we try to integrate our current knowledge on how dynamic instability, regulation of tubulin mass, and self-repair work together to achieve microtubule homeostasis.
Collapse
Affiliation(s)
- Ivana Gasic
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
270
|
Santander VS, Campetelli AN, Monesterolo NE, Rivelli JF, Nigra AD, Arce CA, Casale CH. Tubulin-Na + , K + -ATPase interaction: Involvement in enzymatic regulation and cellular function. J Cell Physiol 2018; 234:7752-7763. [PMID: 30378111 DOI: 10.1002/jcp.27610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/21/2018] [Indexed: 12/22/2022]
Abstract
A new function for tubulin was described by our laboratory: acetylated tubulin forms a complex with Na+ ,K + -ATPase (NKA) and inhibits its activity. This process was shown to be a regulatory factor of physiological importance in cultured cells, human erythrocytes, and several rat tissues. Formation of the acetylated tubulin-NKA complex is reversible. We demonstrated that in cultured cells, high concentrations of glucose induce translocation of acetylated tubulin from cytoplasm to plasma membrane with a consequent inhibition of NKA activity. This effect is reversed by adding glutamate, which is coctransported to the cell with Na + . Another posttranslational modification of tubulin, detyrosinated tubulin, is also involved in the regulation of NKA activity: it enhances the NKA inhibition induced by acetylated tubulin. Manipulation of the content of these modifications of tubulin could work as a new strategy to maintain homeostasis of Na + and K + , and to regulate a variety of functions in which NKA is involved, such as osmotic fragility and deformability of human erythrocytes. The results summarized in this review show that the interaction between tubulin and NKA plays an important role in cellular physiology, both in the regulation of Na + /K + homeostasis and in the rheological properties of the cells, which is mechanically different from other roles reported up to now.
Collapse
Affiliation(s)
- Veronica S Santander
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Alexis N Campetelli
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Noelia E Monesterolo
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Juan F Rivelli
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Ayelen D Nigra
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Carlos A Arce
- entro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba, Argentina
| | - César H Casale
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| |
Collapse
|
271
|
Gu L, Li X, Liu X, Gao M, He Y, Xiong B, Liu H. HDAC3 inhibition disrupts the assembly of meiotic apparatus during porcine oocyte maturation. J Cell Physiol 2018; 234:10178-10183. [DOI: 10.1002/jcp.27687] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Ling Gu
- College of Animal Science & Technology, Nanjing Agricultural University Nanjing China
| | - Xiaoyan Li
- College of Animal Science & Technology, Nanjing Agricultural University Nanjing China
| | - Xiaohui Liu
- College of Animal Science & Technology, Nanjing Agricultural University Nanjing China
| | - Min Gao
- College of Animal Science & Technology, Nanjing Agricultural University Nanjing China
| | - Yongfu He
- College of Animal Science & Technology, Nanjing Agricultural University Nanjing China
| | - Bo Xiong
- College of Animal Science & Technology, Nanjing Agricultural University Nanjing China
| | - Honglin Liu
- College of Animal Science & Technology, Nanjing Agricultural University Nanjing China
| |
Collapse
|
272
|
Pongrakhananon V, Wattanathamsan O, Takeichi M, Chetprayoon P, Chanvorachote P. Loss of CAMSAP3 promotes EMT via the modification of microtubule-Akt machinery. J Cell Sci 2018; 131:jcs.216168. [PMID: 30282632 DOI: 10.1242/jcs.216168] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) plays pivotal roles in a variety of biological processes, including cancer invasion. Although EMT involves alterations of cytoskeletal proteins such as microtubules, the role of microtubules in EMT is not fully understood. Microtubule dynamics are regulated by microtubule-binding proteins, and one such protein is CAMSAP3, which binds the minus-end of microtubules. Here, we show that CAMSAP3 is important to preserve the epithelial phenotypes in lung carcinoma cells. Deletion of CAMSAP3 in human lung carcinoma-derived cell lines showed that CAMSAP3-deficient cells acquired increased mesenchymal features, mostly at the transcriptional level. Analysis of the mechanisms underlying these changes demonstrated that tubulin acetylation was dramatically increased following CAMSAP3 removal, leading to the upregulation of Akt proteins (also known as protein kinase B proteins, hereafter Akt) activity, which is known to promote EMT. These findings suggest that CAMSAP3 functions to protect lung carcinoma cells against EMT by suppressing Akt activity via microtubule regulation and that CAMSAP3 loss promotes EMT in these cells.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Varisa Pongrakhananon
- Cell-Based Drug and Health Product Development Research Unit, Chulalongkorn University, Bangkok 10330, Thailand .,Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Onsurang Wattanathamsan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.,Inter-department Program of Pharmacology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Masatoshi Takeichi
- Laboratory for Cell adhesion and Tissue Patterning, RIKEN Center for Developmental Biology and RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Paninee Chetprayoon
- Nano Safety and Risk Assessment Laboratory, National Nanotechnology Center, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Pithi Chanvorachote
- Cell-Based Drug and Health Product Development Research Unit, Chulalongkorn University, Bangkok 10330, Thailand.,Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
273
|
Caporizzo MA, Chen CY, Salomon AK, Margulies KB, Prosser BL. Microtubules Provide a Viscoelastic Resistance to Myocyte Motion. Biophys J 2018; 115:1796-1807. [PMID: 30322798 DOI: 10.1016/j.bpj.2018.09.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/16/2018] [Accepted: 09/13/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Microtubules (MTs) buckle and bear load during myocyte contraction, a behavior enhanced by post-translational detyrosination. This buckling suggests a spring-like resistance against myocyte shortening, which could store energy and aid myocyte relaxation. Despite this visual suggestion of elastic behavior, the precise mechanical contribution of the cardiac MT network remains to be defined. METHODS Here we experimentally and computationally probe the mechanical contribution of stable MTs and their influence on myocyte function. We use multiple approaches to interrogate viscoelasticity and cell shortening in primary murine myocytes in which either MTs are depolymerized or detyrosination is suppressed and use the results to inform a mathematical model of myocyte viscoelasticity. RESULTS MT ablation by colchicine concurrently enhances both the degree of shortening and speed of relaxation, a finding inconsistent with simple spring-like MT behavior and suggestive of a viscoelastic mechanism. Axial stretch and transverse indentation confirm that MTs increase myocyte viscoelasticity. Specifically, increasing the rate of strain amplifies the MT contribution to myocyte stiffness. Suppressing MT detyrosination with parthenolide or via overexpression of tubulin tyrosine ligase has mechanical consequences that closely resemble colchicine, suggesting that the mechanical impact of MTs relies on a detyrosination-dependent linkage with the myocyte cytoskeleton. Mathematical modeling affirms that alterations in cell shortening conferred by either MT destabilization or tyrosination can be attributed to internal changes in myocyte viscoelasticity. CONCLUSIONS The results suggest that the cardiac MT network regulates contractile amplitudes and kinetics by acting as a cytoskeletal shock-absorber, whereby MTs provide breakable cross-links between the sarcomeric and nonsarcomeric cytoskeleton that resist rapid length changes during both shortening and stretch.
Collapse
Affiliation(s)
- Matthew Alexander Caporizzo
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christina Yingxian Chen
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander Koizumi Salomon
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kenneth B Margulies
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania; Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin L Prosser
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
274
|
Ti SC, Alushin GM, Kapoor TM. Human β-Tubulin Isotypes Can Regulate Microtubule Protofilament Number and Stability. Dev Cell 2018; 47:175-190.e5. [PMID: 30245156 DOI: 10.1016/j.devcel.2018.08.014] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/28/2018] [Accepted: 08/20/2018] [Indexed: 10/28/2022]
Abstract
Cell biological studies have shown that protofilament number, a fundamental feature of microtubules, can correlate with the expression of different tubulin isotypes. However, it is not known if tubulin isotypes directly control this basic microtubule property. Here, we report high-resolution cryo-EM reconstructions (3.5-3.65 Å) of purified human α1B/β3 and α1B/β2B microtubules and find that the β-tubulin isotype can determine protofilament number. Comparisons of atomic models of 13- and 14-protofilament microtubules reveal how tubulin subunit plasticity, manifested in "accordion-like" distributed structural changes, can accommodate distinct lattice organizations. Furthermore, compared to α1B/β3 microtubules, α1B/β2B filaments are more stable to passive disassembly and against depolymerization by MCAK or chTOG, microtubule-associated proteins with distinct mechanisms of action. Mixing tubulin isotypes in different proportions results in microtubules with protofilament numbers and stabilities intermediate to those of isotypically pure filaments. Together, our findings indicate that microtubule protofilament number and stability can be controlled through β-tubulin isotype composition.
Collapse
Affiliation(s)
- Shih-Chieh Ti
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Gregory M Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | - Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
275
|
Abstract
Each neuron forms a single axon and multiple dendrites, and this configuration is important for wiring the brain. How only a single axon extends from a neuron, however, remains unknown. This study demonstrates that CAMSAP3, a protein that binds the minus-end of microtubules, preferentially localizes along axons in hippocampal neurons. Remarkably, mutations of CAMSAP3 lead to production of multiple axons in these neurons. In attempts to uncover mechanisms underlying this abnormal axon extension, the authors found that CAMSAP3-anchored microtubules escape from acetylation, a process mediated by α-tubulin acetyltransferase-1, and depletion of this enzyme abolishes abnormal axon formation in CAMSAP3 mutants. These findings reveal that CAMSAP3 controls microtubule dynamics, preventing tubulin acetylation; this mechanism is required for single-axon formation. The molecular mechanisms that guide each neuron to become polarized, forming a single axon and multiple dendrites, remain unknown. Here we show that CAMSAP3 (calmodulin-regulated spectrin-associated protein 3), a protein that regulates the minus-end dynamics of microtubules, plays a key role in maintaining neuronal polarity. In mouse hippocampal neurons, CAMSAP3 was enriched in axons. Although axonal microtubules were generally acetylated, CAMSAP3 was preferentially localized along a less-acetylated fraction of the microtubules. CAMSAP3-mutated neurons often exhibited supernumerary axons, along with an increased number of neurites having nocodazole-resistant/acetylated microtubules compared with wild-type neurons. Analysis using cell lines showed that CAMSAP3 depletion promoted tubulin acetylation, and conversely, mild overexpression of CAMSAP3 inhibited it, suggesting that CAMSAP3 works to retain nonacetylated microtubules. In contrast, CAMSAP2, a protein related to CAMSAP3, was detected along all neurites, and its loss did not affect neuronal polarity, nor did it cause increased tubulin acetylation. Depletion of α-tubulin acetyltransferase-1 (αTAT1), the key enzyme for tubulin acetylation, abolished CAMSAP3 loss-dependent multiple-axon formation. These observations suggest that CAMSAP3 sustains a nonacetylated pool of microtubules in axons, interfering with the action of αTAT1, and this process is important to maintain neuronal polarity.
Collapse
|
276
|
Fernández-Barrera J, Alonso MA. Coordination of microtubule acetylation and the actin cytoskeleton by formins. Cell Mol Life Sci 2018; 75:3181-3191. [PMID: 29947928 PMCID: PMC11105221 DOI: 10.1007/s00018-018-2855-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/22/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Abstract
The acetylation of the lysine 40 residue of α-tubulin was described more than 30 years ago and has been the subject of intense research ever since. Although the exact function of this covalent modification of tubulin in the cell remains unknown, it has been established that tubulin acetylation confers resilience to mechanical stress on the microtubules. Formins have a dual role in the fate of the actin and tubulin cytoskeletons. On the one hand, they catalyze the formation of actin filaments, and on the other, they bind microtubules, act on their stability, and regulate their acetylation and alignment with actin fibers. Recent evidence indicates that formins coordinate the actin cytoskeleton and tubulin acetylation by modulating the levels of free globular actin (G-actin). G-actin, in turn, controls the activity of the myocardin-related transcription factor-serum response factor transcriptional complex that regulates the expression of the α-tubulin acetyltransferase 1 (α-TAT1) gene, which encodes the main enzyme responsible for tubulin acetylation. The effect of formins on tubulin acetylation is the combined result of their ability to activate α-TAT1 gene transcription and of their capacity to regulate microtubule stabilization. The contribution of these two mechanisms in different formins is discussed, particularly with respect to INF2, a formin that is mutated in hereditary human renal and neurodegenerative disorders.
Collapse
Affiliation(s)
- Jaime Fernández-Barrera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain
| | - Miguel A Alonso
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain.
| |
Collapse
|
277
|
Abstract
Among the different types of cytoskeletal components, microtubules arguably accumulate the greatest diversity of post-translational modifications (PTMs). Acetylation of lysine 40 (K40) of α-tubulin has received particular attention because it is the only tubulin PTM to be found in the lumen of microtubules: most other tubulin PTMs are found at the outer surface of the microtubule. As a consequence, the enzyme catalyzing K40 acetylation needs to penetrate the narrow microtubule lumen to find its substrate. Acetylated microtubules have been considered to be stable, long-lived microtubules; however, until recently, there was little information about whether the longevity of these microtubules is the cause or the consequence of acetylation. Current advances suggest that this PTM helps the microtubule lattice to cope with mechanical stress, thus facilitating microtubule self-repair. These observations now shed new light on the structural integrity of microtubules, as well as on the mechanisms and biological functions of tubulin acetylation. Here, we discuss recent insights into how acetylation is generated in the lumen of microtubules, and how this 'hidden' PTM can control the properties and functions of microtubules.
Collapse
Affiliation(s)
- 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.
| | - Guillaume Montagnac
- Inserm U1170, Gustave Roussy Institute, Université Paris-Saclay, F-94800 Villejuif, France.
| |
Collapse
|
278
|
Bornens M. Cell polarity: having and making sense of direction-on the evolutionary significance of the primary cilium/centrosome organ in Metazoa. Open Biol 2018; 8:180052. [PMID: 30068565 PMCID: PMC6119866 DOI: 10.1098/rsob.180052] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/05/2018] [Indexed: 12/13/2022] Open
Abstract
Cell-autonomous polarity in Metazoans is evolutionarily conserved. I assume that permanent polarity in unicellular eukaryotes is required for cell motion and sensory reception, integration of these two activities being an evolutionarily constrained function. Metazoans are unique in making cohesive multicellular organisms through complete cell divisions. They evolved a primary cilium/centrosome (PC/C) organ, ensuring similar functions to the basal body/flagellum of unicellular eukaryotes, but in different cells, or in the same cell at different moments. The possibility that this innovation contributed to the evolution of individuality, in being instrumental in the early specification of the germ line during development, is further discussed. Then, using the example of highly regenerative organisms like planarians, which have lost PC/C organ in dividing cells, I discuss the possibility that part of the remodelling necessary to reach a new higher-level unit of selection in multi-cellular organisms has been triggered by conflicts among individual cell polarities to reach an organismic polarity. Finally, I briefly consider organisms with a sensorimotor organ like the brain that requires exceedingly elongated polarized cells for its activity. I conclude that beyond critical consequences for embryo development, the conservation of cell-autonomous polarity in Metazoans had far-reaching implications for the evolution of individuality.
Collapse
Affiliation(s)
- Michel Bornens
- Institut Curie, PSL Research University, CNRS - UMR 144, 75005 Paris, France
| |
Collapse
|
279
|
Cilium structure, assembly, and disassembly regulated by the cytoskeleton. Biochem J 2018; 475:2329-2353. [PMID: 30064990 PMCID: PMC6068341 DOI: 10.1042/bcj20170453] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 12/17/2022]
Abstract
The cilium, once considered a vestigial structure, is a conserved, microtubule-based organelle critical for transducing extracellular chemical and mechanical signals that control cell polarity, differentiation, and proliferation. The cilium undergoes cycles of assembly and disassembly that are controlled by complex inter-relationships with the cytoskeleton. Microtubules form the core of the cilium, the axoneme, and are regulated by post-translational modifications, associated proteins, and microtubule dynamics. Although actin and septin cytoskeletons are not major components of the axoneme, they also regulate cilium organization and assembly state. Here, we discuss recent advances on how these different cytoskeletal systems affect cilium function, structure, and organization.
Collapse
|
280
|
Yip TF, Selim ASM, Lian I, Lee SMY. Advancements in Host-Based Interventions for Influenza Treatment. Front Immunol 2018; 9:1547. [PMID: 30042762 PMCID: PMC6048202 DOI: 10.3389/fimmu.2018.01547] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/22/2018] [Indexed: 12/15/2022] Open
Abstract
Influenza is a major acute respiratory infection that causes mortality and morbidity worldwide. Two classes of conventional antivirals, M2 ion channel blockers and neuraminidase inhibitors, are mainstays in managing influenza disease to lessen symptoms while minimizing hospitalization and death in patients with severe influenza. However, the development of viral resistance to both drug classes has become a major public health concern. Vaccines are prophylaxis mainstays but are limited in efficacy due to the difficulty in matching predicted dominant viral strains to circulating strains. As such, other potential interventions are being explored. Since viruses rely on host cellular functions to replicate, recent therapeutic developments focus on targeting host factors involved in virus replication. Besides controlling virus replication, potential targets for drug development include controlling virus-induced host immune responses such as the recently suggested involvement of innate lymphoid cells and NADPH oxidases in influenza virus pathogenesis and immune cell metabolism. In this review, we will discuss the advancements in novel host-based interventions for treating influenza disease.
Collapse
Affiliation(s)
- Tsz-Fung Yip
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| | - Aisha Sami Mohammed Selim
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ida Lian
- School of Life Sciences and Chemical Technology, Ngee Ann Polytechnic, Singapore, Singapore
| | - Suki Man-Yan Lee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
281
|
The role of tubulin-tubulin lattice contacts in the mechanism of microtubule dynamic instability. Nat Struct Mol Biol 2018; 25:607-615. [PMID: 29967541 PMCID: PMC6201834 DOI: 10.1038/s41594-018-0087-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/01/2018] [Indexed: 11/08/2022]
Abstract
Microtubules form from longitudinally and laterally assembling tubulin α-β dimers. The assembly induces strain in tubulin, resulting in cycles of microtubule catastrophe and regrowth. This 'dynamic instability' is governed by GTP hydrolysis that renders the microtubule lattice unstable, but it is unclear how. We used a human microtubule nucleating and stabilizing neuronal protein, doublecortin, and high-resolution cryo-EM to capture tubulin's elusive hydrolysis intermediate GDP•Pi state, alongside the prehydrolysis analog GMPCPP state and the posthydrolysis GDP state with and without an anticancer drug, Taxol. GTP hydrolysis to GDP•Pi followed by Pi release constitutes two distinct structural transitions, causing unevenly distributed compressions of tubulin dimers, thereby tightening longitudinal and loosening lateral interdimer contacts. We conclude that microtubule catastrophe is triggered because the lateral contacts can no longer counteract the strain energy stored in the lattice, while reinforcement of the longitudinal contacts may support generation of force.
Collapse
|
282
|
Abstract
Microtubules are dynamic polymers of αβ-tubulin that are essential for intracellular organization, organelle trafficking and chromosome segregation. Microtubule growth and shrinkage occur via addition and loss of αβ-tubulin subunits, which are biochemical processes. Dynamic microtubules can also engage in mechanical processes, such as exerting forces by pushing or pulling against a load. Recent advances at the intersection of biochemistry and mechanics have revealed the existence of multiple conformations of αβ-tubulin subunits and their central role in dictating the mechanisms of microtubule dynamics and force generation. It has become apparent that microtubule-associated proteins (MAPs) selectively target specific tubulin conformations to regulate microtubule dynamics, and mechanical forces can also influence microtubule dynamics by altering the balance of tubulin conformations. Importantly, the conformational states of tubulin dimers are likely to be coupled throughout the lattice: the conformation of one dimer can influence the conformation of its nearest neighbours, and this effect can propagate over longer distances. This coupling provides a long-range mechanism by which MAPs and forces can modulate microtubule growth and shrinkage. These findings provide evidence that the interplay between biochemistry and mechanics is essential for the cellular functions of microtubules.
Collapse
Affiliation(s)
- Gary J Brouhard
- Department of Biology, McGill University, Montréal, Quebec, Canada.
| | - Luke M Rice
- Department of Biophysics, University of Texas Southwestern, Dallas, TX, USA.
| |
Collapse
|
283
|
Weng RR, Yang TT, Huang CE, Chang CW, Wang WJ, Liao JC. Super-Resolution Imaging Reveals TCTN2 Depletion-Induced IFT88 Lumen Leakage and Ciliary Weakening. Biophys J 2018; 115:263-275. [PMID: 29866362 DOI: 10.1016/j.bpj.2018.04.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/07/2018] [Accepted: 04/23/2018] [Indexed: 11/28/2022] Open
Abstract
The primary cilium is an essential organelle mediating key signaling activities, such as sonic hedgehog signaling. The molecular composition of the ciliary compartment is distinct from that of the cytosol, with the transition zone (TZ) gated the ciliary base. The TZ is a packed and organized protein complex containing multiple ciliopathy-associated protein species. Tectonic 2 (TCTN2) is one of the TZ proteins in the vicinity of the ciliary membrane, and its mutation is associated with Meckel syndrome. Despite its importance in ciliopathies, the role of TCTN2 in ciliary structure and molecules remains unclear. Here, we created a CRISPR/Cas9 TCTN2 knockout human retinal pigment epithelial cell line and conducted quantitative analysis of geometric localization using both wide-field and super-resolution microscopy techniques. We found that TCTN2 depletion resulted in partial TZ damage, loss of ciliary membrane proteins, leakage of intraflagellar transport protein IFT88 toward the basal body lumen, and cilium shortening and curving. The basal body lumen occupancy of IFT88 was also observed in si-RPGRIP1L cells and cytochalasin-D-treated wild-type cells, suggesting varying lumen accessibility for intraflagellar transport proteins under different perturbed conditions. Our findings support two possible models for the lumen leakage of IFT88, i.e., a tip leakage model and a misregulation model. Together, our quantitative image analysis augmented by super-resolution microscopy facilitates the observation of structural destruction and molecular redistribution in TCTN2-/- cilia, shedding light on mechanistic understanding of TZ-protein-associated ciliopathies.
Collapse
Affiliation(s)
- Rueyhung Roc Weng
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - T Tony Yang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-En Huang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-Wei Chang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Won-Jing Wang
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei, Taiwan
| | - Jung-Chi Liao
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan; Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
284
|
Procter DJ, Banerjee A, Nukui M, Kruse K, Gaponenko V, Murphy EA, Komarova Y, Walsh D. The HCMV Assembly Compartment Is a Dynamic Golgi-Derived MTOC that Controls Nuclear Rotation and Virus Spread. Dev Cell 2018; 45:83-100.e7. [PMID: 29634939 DOI: 10.1016/j.devcel.2018.03.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 02/08/2018] [Accepted: 03/14/2018] [Indexed: 10/17/2022]
Abstract
Human cytomegalovirus (HCMV), a leading cause of congenital birth defects, forms an unusual cytoplasmic virion maturation site termed the "assembly compartment" (AC). Here, we show that the AC also acts as a microtubule-organizing center (MTOC) wherein centrosome activity is suppressed and Golgi-based microtubule (MT) nucleation is enhanced. This involved viral manipulation of discrete functions of MT plus-end-binding (EB) proteins. In particular, EB3, but not EB1 or EB2, was recruited to the AC and was required to nucleate MTs that were rapidly acetylated. EB3-regulated acetylated MTs were necessary for nuclear rotation prior to cell migration, maintenance of AC structure, and optimal virus replication. Independently, a myristoylated peptide that blocked EB3-mediated enrichment of MT regulatory proteins at Golgi regions of the AC also suppressed acetylated MT formation, nuclear rotation, and infection. Thus, HCMV offers new insights into the regulation and functions of Golgi-derived MTs and the therapeutic potential of targeting EB3.
Collapse
Affiliation(s)
- Dean J Procter
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Avik Banerjee
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Masatoshi Nukui
- Department of Translational Medicine, Baruch S. Blumberg Research Institute, Doylestown, PA 18902, USA; Forge Life Science, Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA
| | - Kevin Kruse
- Department of Pharmacology and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Eain A Murphy
- Department of Translational Medicine, Baruch S. Blumberg Research Institute, Doylestown, PA 18902, USA; Forge Life Science, Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA
| | - Yulia Komarova
- Department of Pharmacology and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Derek Walsh
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
285
|
Zhong X, Lee HN, Kim SH, Park SA, Kim W, Cha YN, Surh YJ. Myc-nick promotes efferocytosis through M2 macrophage polarization during resolution of inflammation. FASEB J 2018; 32:5312-5325. [PMID: 29718706 DOI: 10.1096/fj.201800223r] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A key event required for effective resolution of inflammation is efferocytosis, which is defined as phagocytic removal of apoptotic cells mostly by macrophages acquiring an alternatively activated phenotype (M2). c-Myc has been reported to play a role in alternative activation of human macrophages and is proposed as one of the M2 macrophage markers. We found that M2-like peritoneal macrophages from zymosan A-treated mice exhibited a marked accumulation of Myc-nick, a truncated protein generated by a Calpain-mediated proteolytic cleavage of full-length c-Myc. Further, ectopic expression of Myc-nick in murine bone marrow-derived macrophages promoted the M2 polarization and, consequently, enhanced their efferocytic capability. Notably, Myc-nick-induced efferocytosis was found to be tightly associated with α-tubulin acetylation by K acetyltransferase 2a (Kat2a/Gcn5) activity. These findings suggest Myc-nick as a novel proresolving mediator that has a fundamental function in maintaining homeostasis under inflammatory conditions.-Zhong, X., Lee, H.-N., Kim, S. H., Park, S.-A., Kim, W., Cha, Y.-N., Surh, Y.-J. Myc-nick promotes efferocytosis through M2 macrophage polarization during resolution of inflammation.
Collapse
Affiliation(s)
- Xiancai Zhong
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Ha-Na Lee
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Seung Hyeon Kim
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| | - Sin-Aye Park
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Wonki Kim
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Young-Nam Cha
- Department of Pharmacology and Toxicology, College of Medicine, Inha University, Incheon, South Korea; and
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
286
|
TAK1 activation of alpha-TAT1 and microtubule hyperacetylation control AKT signaling and cell growth. Nat Commun 2018; 9:1696. [PMID: 29703898 PMCID: PMC5923212 DOI: 10.1038/s41467-018-04121-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 04/04/2018] [Indexed: 01/04/2023] Open
Abstract
Acetylation of microtubules (MT) confers mechanical stability necessary for numerous functions including cell cycle and intracellular transport. Although αTAT1 is a major MT acetyltransferase, how this enzyme is regulated remains much less clear. Here we report TGF-β-activated kinase 1 (TAK1) as a key activator of αTAT1. TAK1 directly interacts with and phosphorylates αTAT1 at Ser237 to critically enhance its catalytic activity, as mutating this site to alanine abrogates, whereas a phosphomimetic induces MT hyperacetylation across cell types. Using a custom phospho-αTAT1-Ser237 antibody, we screen various mouse tissues to discover that brain contains some of the highest TAK1-dependent αTAT1 activity, which, accordingly, is diminished rapidly upon intra-cerebral injection of a TAK1 inhibitor. Lastly, we show that TAK1 selectively inhibits AKT to suppress mitogenic and metabolism-related pathways through MT-based mechanisms in culture and in vivo. Collectively, our findings support a fundamental new role for TGF-β signaling in MT-related functions and disease. Acetylation of microtubules (MT) confers mechanical stability necessary for numerous cellular functions but its regulation is unclear. Here the authors show that the MT acetyltransferase αTAT1 is regulated by TGF-β-activated kinase 1 implicating TGF-β signaling in MT-related functions and disease.
Collapse
|
287
|
Serra E, Succu S, Berlinguer F, Porcu C, Leoni GG, Naitana S, Gadau SD. Tubulin posttranslational modifications in in vitro matured prepubertal and adult ovine oocytes. Theriogenology 2018; 114:237-243. [PMID: 29660626 DOI: 10.1016/j.theriogenology.2018.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/09/2018] [Accepted: 04/05/2018] [Indexed: 11/28/2022]
Abstract
Microtubules (MTs), polymers of alpha/beta-tubulin heterodimers, are involved in crucial functions in eukaryotic cells. MTs physiology can be influenced by a variety of post-translational modifications (PTMs), including tyrosination, detyrosination, delta 2 modification, acetylation, polyglutamylation, polyglycylation. In mammalian oocytes, MTs are essential for meiosis, regulating the formation of meiotic spindle and chromosomes movements. Considering that the patterns of tubulin PTMs (tyrosination, detyrosination, acetylation, polyglutamylation and delta 2 modification) have not been investigated in ovine oocytes, this study has been designed to investigate their presence and quantification in in vitro matured (IVM) adult and prepubertal ovine oocytes. Oocytes from adult and lamb Sarda ewes, regularly slaughtered at the local abattoir, were in vitro matured, fixed, and processed by indirect immunofluorescence and confocal microscopy analyses at metaphase II stage. Our results revealed a well detectable signal for total, tyrosinated and acetylated α-tubulin in meiotic spindle of both sheep and lamb oocytes. On the other hand, no immunopositivity were appreciable for detyrosinated, polyglutamylated, and delta 2 tubulin in meiotic spindle of both sheep and lamb oocytes. As regard the tyrosinated and the acetylated α-tubulin PTMs, through the quantification of the fluorescence intensity, we did not find significant differences in their expression in meiotic spindle of sheep, while in lamb the acetylated tubulin levels were predominant in comparison with tyrosinated. Our results in addition to investigating for the first time the different tubulin PTMs in the spindle organization of ovine oocytes, showed a different microtubule pattern between adult and prepubertal oocytes. The microtubule cytoskeleton survey may thus suggest further cues to better understand skill-related problems in in the acquisition of oocyte competence.
Collapse
Affiliation(s)
- E Serra
- Department of Veterinary Medicine, University of Sassari, Italy
| | - S Succu
- Department of Veterinary Medicine, University of Sassari, Italy
| | - F Berlinguer
- Department of Veterinary Medicine, University of Sassari, Italy
| | - C Porcu
- Department of Veterinary Medicine, University of Sassari, Italy
| | - G G Leoni
- Department of Veterinary Medicine, University of Sassari, Italy
| | - S Naitana
- Department of Veterinary Medicine, University of Sassari, Italy
| | - S D Gadau
- Department of Veterinary Medicine, University of Sassari, Italy.
| |
Collapse
|
288
|
Mondal P, Das G, Khan J, Pradhan K, Ghosh S. Crafting of Neuroprotective Octapeptide from Taxol-Binding Pocket of β-Tubulin. ACS Chem Neurosci 2018; 9:615-625. [PMID: 29155559 DOI: 10.1021/acschemneuro.7b00457] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Microtubules play a crucial role in maintaining the shape and function of neurons. During progression of Alzheimer's disease (AD), severe destabilization of microtubules occurs, which leads to the permanent disruption of signal transduction processes and memory loss. Thus, microtubule stabilization is one of the key requirements for the treatment of AD. Taxol, a microtubule stabilizing anticancer drug, has been considered as a potential anti-AD drug but was never tested in AD patients, likely because of its' toxic nature and poor brain exposure. However, other microtubule-targeting agents such as epothilone D (BMS-241027) and TPI-287 (abeotaxane) and NAP peptide (davunetide) have entered in AD clinical programs. Therefore, the taxol binding pocket of tubulin could be a potential site for designing of mild and noncytotoxic microtubule stabilizing molecules. Here, we adopted an innovative strategy for the development of a peptide based microtubule stabilizer, considering the taxol binding pocket of β-tubulin, by using alanine scanning mutagenesis technique. This approach lead us to a potential octapeptide, which strongly binds to the taxol pocket of β-tubulin, serves as an excellent microtubule stabilizer, increases the expression of acetylated tubulin, and acts as an Aβ aggregation inhibitor and neuroprotective agent. Further, results revealed that this peptide is nontoxic against both PC12 derived neurons and primary cortical neurons. We believe that our strategy and discovery of peptide-based microtubule stabilizer will open the door for the development of potential anti-AD therapeutics in near future.
Collapse
Affiliation(s)
- Prasenjit Mondal
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Gaurav Das
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Juhee Khan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Krishnangsu Pradhan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
| | - Surajit Ghosh
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| |
Collapse
|
289
|
Aher A, Akhmanova A. Tipping microtubule dynamics, one protofilament at a time. Curr Opin Cell Biol 2018; 50:86-93. [PMID: 29573640 DOI: 10.1016/j.ceb.2018.02.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 02/19/2018] [Accepted: 02/28/2018] [Indexed: 01/10/2023]
Abstract
Microtubules are polymeric tubes that switch between phases of growth and shortening, and this property is essential to drive key cellular processes. Microtubules are composed of protofilaments formed by longitudinally arranged tubulin dimers. Microtubule dynamics can be affected by structural perturbations at the plus end, such as end tapering, and targeting only a small subset of protofilaments can alter the dynamics of the whole microtubule. Microtubule lattice plasticity, including compaction along the longitudinal axis upon GTP hydrolysis and tubulin dimer loss and reinsertion along microtubule shafts can also affect microtubule dynamics or mechanics. Microtubule behaviour can be fine-tuned by post-translational modifications and tubulin isotypes, which together support the diversity of microtubule functions within and across various cell types or cell cycle and developmental stages.
Collapse
Affiliation(s)
- Amol Aher
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
290
|
The tubulin code in neuronal polarity. Curr Opin Neurobiol 2018; 51:95-102. [PMID: 29554585 DOI: 10.1016/j.conb.2018.03.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 11/22/2022]
Abstract
Cells depend on the asymmetric distribution of their components for homeostasis, differentiation and movement. In no other cell type is this requirement more critical than in the neuron where complex structures are generated during process growth and elaboration and cargo is transported over distances several thousand times the cell body diameter. Microtubules act both as dynamic structural elements and as tracks for intracellular transport. Microtubules are mosaic polymers containing multiple tubulin isoforms functionalized with abundant posttranslational modifications that are asymmetrically distributed in neurons. An increasing body of evidence supports the hypothesis that the combinatorial information expressed through tubulin genetic and chemical diversity controls microtubule dynamics, mechanics and interactions with microtubule effectors and thus constitutes a 'tubulin code'. Here we give a brief overview of tubulin isoform usage and posttranslational modifications in the neuron, and highlight recent progress in understanding the molecular mechanisms of the tubulin code.
Collapse
|
291
|
Blanquie O, Bradke F. Cytoskeleton dynamics in axon regeneration. Curr Opin Neurobiol 2018; 51:60-69. [PMID: 29544200 DOI: 10.1016/j.conb.2018.02.024] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/22/2018] [Accepted: 02/26/2018] [Indexed: 11/19/2022]
Abstract
Recent years have seen cytoskeleton dynamics emerging as a key player in axon regeneration. The cytoskeleton, in particular microtubules and actin, ensures the growth of neuronal processes and maintains the singular, highly polarized shape of neurons. Following injury, adult central axons are tipped by a dystrophic structure, the retraction bulb, which prevents their regeneration. Abnormal cytoskeleton dynamics are responsible for the formation of this growth-incompetent structure but pharmacologically modulating cytoskeleton dynamics of injured axons can transform this structure into a growth-competent growth cone. The cytoskeleton also drives the migration of scar-forming cells after an injury. Targeting its dynamics modifies the composition of the inhibitory environment formed by scar tissue and renders it more permissive for regenerating axons. Hence, cytoskeleton dynamics represent an appealing target to promote axon regeneration. As some of cytoskeleton-targeting drugs are used in the clinics for other purposes, they hold the promise to be used as a basis for a regenerative therapy after a spinal cord injury.
Collapse
Affiliation(s)
- Oriane Blanquie
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
| | - Frank Bradke
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
| |
Collapse
|
292
|
Yu J, Lai C, Shim H, Xie C, Sun L, Long CX, Ding J, Li Y, Cai H. Genetic ablation of dynactin p150 Glued in postnatal neurons causes preferential degeneration of spinal motor neurons in aged mice. Mol Neurodegener 2018; 13:10. [PMID: 29490687 PMCID: PMC5831668 DOI: 10.1186/s13024-018-0242-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/19/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Dynactin p150Glued, the largest subunit of the dynactin macromolecular complex, binds to both microtubules and tubulin dimers through the N-terminal cytoskeleton-associated protein and glycine-rich (CAP-Gly) and basic domains, and serves as an anti-catastrophe factor in stabilizing microtubules in neurons. P150Glued also initiates dynein-mediated axonal retrograde transport. Multiple missense mutations at the CAP-Gly domain of p150Glued are associated with motor neuron diseases and other neurodegenerative disorders, further supporting the importance of microtubule domains (MTBDs) in p150Glued functions. However, most functional studies were performed in vitro. Whether p150Glued is required for neuronal function and survival in vivo is unknown. METHODS Using Cre-loxP genetic manipulation, we first generated a line of p150Glued knock-in mice by inserting two LoxP sites flanking the MTBD-coding exons 2 to 4 of p150Glued-encoding Dctn1 gene (Dctn1LoxP/), and then crossbred the resulting Dctn1LoxP/ mice with Thy1-Cre mice to generate the bigenic p150Glued (Dctn1LoxP/LoxP; Thy1-Cre) conditional knockout (cKO) mice for the downstream motor behavioral and neuropathological studies. RESULTS P150Glued expression was completely abolished in Cre-expressing postnatal neurons, including corticospinal motor neurons (CSMNs) and spinal motor neurons (SMNs), while the MTBD-truncated forms remained. P150Glued ablation did not affect the formation of dynein/dynactin complex in neurons. The p150Glued cKO mice did not show any obvious developmental phenotypes, but exhibited impairments in motor coordination and rearing after 12 months of age. Around 20% loss of SMNs was found in the lumbar spinal cord of 18-month-old cKO mice, in company with increased gliosis, neuromuscular junction (NMJ) disintegration and muscle atrophy. By contrast, no obvious degeneration of CSMNs, striatal neurons, midbrain dopaminergic neurons, cerebellar granule cells or Purkinje cells was observed. Abnormal accumulation of acetylated α-tubulin, and autophagosome/lysosome proteins was found in the SMNs of aged cKO mice. Additionally, the total and cell surface levels of glutamate receptors were also substantially elevated in the p150Glued-depleted spinal neurons, in correlation with increased vulnerability to excitotoxicity. CONCLUSION Overall, our findings demonstrate that p150Glued is particularly required to maintain the function and survival of SMNs during aging. P150Glued may exert its protective function through regulating the transportation of autophagosomes, lysosomes, and postsynaptic glutamate receptors in neurons.
Collapse
Affiliation(s)
- Jia Yu
- Institute for Geriatrics and Rehabilitation, Beijing Geriatric Hospital, Beijing University of Chinese Medicine, Beijing, 100095, People's Republic of China
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A112, MSC 3707, 35 Convent Drive, Bethesda, MD, 20892-3707, USA
| | - Chen Lai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A112, MSC 3707, 35 Convent Drive, Bethesda, MD, 20892-3707, USA
- Present address: Symptom Management Branch, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hoon Shim
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A112, MSC 3707, 35 Convent Drive, Bethesda, MD, 20892-3707, USA
- Department of Anesthesiology, Albert Einstein College of Medicine, Bronx, New York, 10467, USA
| | - Chengsong Xie
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A112, MSC 3707, 35 Convent Drive, Bethesda, MD, 20892-3707, USA
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A112, MSC 3707, 35 Convent Drive, Bethesda, MD, 20892-3707, USA
| | - Cai-Xia Long
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A112, MSC 3707, 35 Convent Drive, Bethesda, MD, 20892-3707, USA
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jinhui Ding
- Computational Biology Core, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yan Li
- NINDS Protein/Peptide Sequencing Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A112, MSC 3707, 35 Convent Drive, Bethesda, MD, 20892-3707, USA.
| |
Collapse
|
293
|
Cep120 promotes microtubule formation through a unique tubulin binding C2 domain. J Struct Biol 2018; 203:62-70. [PMID: 29398280 DOI: 10.1016/j.jsb.2018.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 02/05/2023]
Abstract
Centrioles are microtubule-based structures that play essential roles in cell division and cilia biogenesis. Cep120 is an important protein for correct centriole formation and mutations in the Cep120 gene cause severe human diseases like Joubert syndrome and complex ciliopathies. Here, we show that Cep120 contains three consecutive C2 domains that are followed by a coiled-coil dimerization domain. Surprisingly, unlike the classical C2 domains, all three Cep120 C2 domains lack calcium- and phospholipid-binding activities. However, biophysical and biochemical assays revealed that the N-terminal Cep120 C2 domain (C2A) binds to both tubulin and microtubules, and promotes microtubule formation. Structural analyses coupled with mutagenesis identified a highly conserved, positively charged residue patch on the surface of Cep120 C2A, which mediates the interaction with tubulin and microtubules. Together, our results establish Cep120 C2A as a unique microtubule-binding domain. They further provide insights into the molecular mechanism of Cep120 during centriole biogenesis.
Collapse
|
294
|
Aqueous extract of Triphala inhibits cancer cell proliferation through perturbation of microtubule assembly dynamics. Biomed Pharmacother 2018; 98:76-81. [DOI: 10.1016/j.biopha.2017.12.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/23/2017] [Accepted: 12/04/2017] [Indexed: 01/20/2023] Open
|
295
|
Howes SC, Geyer EA, LaFrance B, Zhang R, Kellogg EH, Westermann S, Rice LM, Nogales E. Structural and functional differences between porcine brain and budding yeast microtubules. Cell Cycle 2018; 17:278-287. [PMID: 29278985 PMCID: PMC5914886 DOI: 10.1080/15384101.2017.1415680] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The cytoskeleton of eukaryotic cells relies on microtubules to perform many essential functions. We have previously shown that, in spite of the overall conservation in sequence and structure of tubulin subunits across species, there are differences between mammalian and budding yeast microtubules with likely functional consequences for the cell. Here we expand our structural and function comparison of yeast and porcine microtubules to show different distribution of protofilament number in microtubules assembled in vitro from these two species. The different geometry at lateral contacts between protofilaments is likely due to a more polar interface in yeast. We also find that yeast tubulin forms longer and less curved oligomers in solution, suggesting stronger tubulin:tubulin interactions along the protofilament. Finally, we observed species-specific plus-end tracking activity for EB proteins: yeast Bim1 tracked yeast but not mammalian MTs, and human EB1 tracked mammalian but not yeast MTs. These findings further demonstrate that subtle sequence differences in tubulin sequence can have significant structural and functional consequences in microtubule structure and behavior.
Collapse
Affiliation(s)
- Stuart C Howes
- a Biophysics Graduate Group , UC Berkeley , CA 94720 , USA and Department of Molecular Cell Biology , Leiden University Medical Center , 2333 ZC Leiden , Netherlands
| | - Elisabeth A Geyer
- b UT Southwestern Medical Center , Departments of Biophysics and Biochemistry , Dallas , TX 75390 , USA
| | - Benjamin LaFrance
- c Molecular and Cell Biology Graduate Program , UC Berkeley , CA 94720 , USA
| | - Rui Zhang
- d Molecular Biophysics and Integrated Bioimaging , Lawrence Berkeley National Laboratory , CA 94720 , USA.,e Howard Hughes Medical Institute , UC Berkeley , CA 94720-3220 , USA.,f Department of Biochemistry and Molecular Biophysics , Washington University School of Medicine , St. Louis , MO 63110 , USA
| | - Elizabeth H Kellogg
- d Molecular Biophysics and Integrated Bioimaging , Lawrence Berkeley National Laboratory , CA 94720 , USA.,e Howard Hughes Medical Institute , UC Berkeley , CA 94720-3220 , USA
| | - Stefan Westermann
- g Research Institute of Molecular Pathology , Dr. Bohr-Gasse 7, 1030 Vienna , Austria
| | - Luke M Rice
- b UT Southwestern Medical Center , Departments of Biophysics and Biochemistry , Dallas , TX 75390 , USA
| | - Eva Nogales
- d Molecular Biophysics and Integrated Bioimaging , Lawrence Berkeley National Laboratory , CA 94720 , USA.,e Howard Hughes Medical Institute , UC Berkeley , CA 94720-3220 , USA.,h Molecular and Cell Biology Department and QB3 Institute , UC Berkeley , CA 94720 , USA
| |
Collapse
|
296
|
Fernández-Barrera J, Bernabé-Rubio M, Casares-Arias J, Rangel L, Fernández-Martín L, Correas I, Alonso MA. The actin-MRTF-SRF transcriptional circuit controls tubulin acetylation via α-TAT1 gene expression. J Cell Biol 2018; 217:929-944. [PMID: 29321169 PMCID: PMC5839776 DOI: 10.1083/jcb.201702157] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/28/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023] Open
Abstract
The role of formins in microtubules is not well understood. In this study, we have investigated the mechanism by which INF2, a formin mutated in degenerative renal and neurological hereditary disorders, controls microtubule acetylation. We found that silencing of INF2 in epithelial RPE-1 cells produced a dramatic drop in tubulin acetylation, increased the G-actin/F-actin ratio, and impaired myocardin-related transcription factor (MRTF)/serum response factor (SRF)-dependent transcription, which is known to be repressed by increased levels of G-actin. The effect on tubulin acetylation was caused by the almost complete absence of α-tubulin acetyltransferase 1 (α-TAT1) messenger RNA (mRNA). Activation of the MRTF-SRF transcriptional complex restored α-TAT1 mRNA levels and tubulin acetylation. Several functional MRTF-SRF-responsive elements were consistently identified in the α-TAT1 gene. The effect of INF2 silencing on microtubule acetylation was also observed in epithelial ECV304 cells, but not in Jurkat T cells. Therefore, the actin-MRTF-SRF circuit controls α-TAT1 transcription. INF2 regulates the circuit, and hence microtubule acetylation, in cell types where it has a prominent role in actin polymerization.
Collapse
Affiliation(s)
- Jaime Fernández-Barrera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel Bernabé-Rubio
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Casares-Arias
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Rangel
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.,Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Fernández-Martín
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Isabel Correas
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.,Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel A Alonso
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
297
|
Prystopiuk V, Fels B, Simon CS, Liashkovich I, Pasrednik D, Kronlage C, Wedlich-Söldner R, Oberleithner H, Fels J. A two-phase response of endothelial cells to hydrostatic pressure. J Cell Sci 2018; 131:jcs.206920. [DOI: 10.1242/jcs.206920] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 05/10/2018] [Indexed: 01/15/2023] Open
Abstract
The vascular endothelium is exposed to three types of mechanical forces: blood flow-mediated shear stress, vessel-diameter dependent wall tension and hydrostatic pressure. Despite considerable variations of blood pressure in normal and pathological physiology, little is known about the acute molecular and cellular effects of hydrostatic pressure on endothelial cells. Here, we used a combination of quantitative fluorescence microscopy, atomic force microscopy and molecular perturbations to characterize the specific response of endothelial cells to pressure application. We identified a two-phase response of endothelial cells to acute (1 h) vs. chronic (24 h) pressure application (100 mmHg). While both regimes induce cortical stiffening, the acute response is linked to calcium-mediated myosin activation, whereas the chronic cell response is dominated by increased cortical actin density and a loss in endothelial barrier function. GsMTx-4 and amiloride inhibit the acute pressure response, which suggest the sodium channel ENaC as key player in endothelial pressure sensing. The described two-phase pressure response may participate in the differential effects of transient changes in blood pressure and hypertension.
Collapse
Affiliation(s)
- Valeria Prystopiuk
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
- current address: Institute of Life Sciences, Université Catholique de Louvain, Croix du Sud, 4-5, bte L7.07.06, Louvain-la-Neuve B-1348, Belgium
| | - Benedikt Fels
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Caroline Sophie Simon
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Dzmitry Pasrednik
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Cornelius Kronlage
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Roland Wedlich-Söldner
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Hans Oberleithner
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Johannes Fels
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| |
Collapse
|
298
|
Balabanian L, Berger CL, Hendricks AG. Acetylated Microtubules Are Preferentially Bundled Leading to Enhanced Kinesin-1 Motility. Biophys J 2017; 113:1551-1560. [PMID: 28978447 DOI: 10.1016/j.bpj.2017.08.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022] Open
Abstract
The motor proteins kinesin and dynein transport organelles, mRNA, proteins, and signaling molecules along the microtubule cytoskeleton. In addition to serving as tracks for transport, the microtubule cytoskeleton directs intracellular trafficking by regulating the activity of motor proteins through the organization of the filament network, microtubule-associated proteins, and tubulin posttranslational modifications. However, it is not well understood how these factors influence motor motility, and in vitro assays and live cell observations often produce disparate results. To systematically examine the factors that contribute to cytoskeleton-based regulation of motor protein motility, we extracted intact microtubule networks from cells and tracked the motility of single fluorescently labeled motor proteins on these cytoskeletons. We find that tubulin acetylation alone does not directly affect kinesin-1 motility. However, acetylated microtubules are predominantly bundled, and bundling enhances kinesin run lengths and provides a greater number of available kinesin binding sites. The neuronal MAP tau is also not sensitive to tubulin acetylation, but enriches preferentially on highly curved regions of microtubules where it strongly inhibits kinesin motility. Taken together, these results suggest that the organization of the microtubule network is a key contributor to the regulation of motor-based transport.
Collapse
Affiliation(s)
- Linda Balabanian
- Department of Bioengineering, McGill University, Montreal, Québec, Canada
| | - Christopher L Berger
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont
| | - Adam G Hendricks
- Department of Bioengineering, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
299
|
Xu Z, Schaedel L, Portran D, Aguilar A, Gaillard J, Marinkovich MP, Théry M, Nachury MV. Microtubules acquire resistance from mechanical breakage through intralumenal acetylation. Science 2017; 356:328-332. [PMID: 28428427 DOI: 10.1126/science.aai8764] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 03/24/2017] [Indexed: 12/11/2022]
Abstract
Eukaryotic cells rely on long-lived microtubules for intracellular transport and as compression-bearing elements. We considered that long-lived microtubules are acetylated inside their lumen and that microtubule acetylation may modify microtubule mechanics. Here, we found that tubulin acetylation is required for the mechanical stabilization of long-lived microtubules in cells. Depletion of the tubulin acetyltransferase TAT1 led to a significant increase in the frequency of microtubule breakage. Nocodazole-resistant microtubules lost upon removal of acetylation were largely restored by either pharmacological or physical removal of compressive forces. In in vitro reconstitution experiments, acetylation was sufficient to protect microtubules from mechanical breakage. Thus, acetylation increases mechanical resilience to ensure the persistence of long-lived microtubules.
Collapse
Affiliation(s)
- Zhenjie Xu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. .,Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305-5168, USA
| | - Laura Schaedel
- CytoMorpho Laboratory, Laboratory of Cell and Plant Physiology (LPCV), UMR 5168, Biosciences and Biotechnology Institute of Grenoble, CEA/INRA/CNRS/Université Grenoble-Alpes, 17 rue des Martyrs, 38054 Grenoble, France
| | - Didier Portran
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Andrea Aguilar
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Jérémie Gaillard
- CytoMorpho Laboratory, Laboratory of Cell and Plant Physiology (LPCV), UMR 5168, Biosciences and Biotechnology Institute of Grenoble, CEA/INRA/CNRS/Université Grenoble-Alpes, 17 rue des Martyrs, 38054 Grenoble, France
| | - M Peter Marinkovich
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305-5168, USA.,Division of Dermatology, Palo Alto Veterans Affairs Medical Center, Palo Alto, CA 94305, USA
| | - Manuel Théry
- CytoMorpho Laboratory, Laboratory of Cell and Plant Physiology (LPCV), UMR 5168, Biosciences and Biotechnology Institute of Grenoble, CEA/INRA/CNRS/Université Grenoble-Alpes, 17 rue des Martyrs, 38054 Grenoble, France.,CytoMorpho Laboratory, A2T, UMRS 1160, Institut Universitaire d'Hématologie, Hôpital Saint Louis, INSERM/AP-HP/Université Paris Diderot, 1 Avenue Claude Vellefaux, 75010 Paris, France
| | - Maxence V Nachury
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA.
| |
Collapse
|
300
|
EB1-binding-myomegalin protein complex promotes centrosomal microtubules functions. Proc Natl Acad Sci U S A 2017; 114:E10687-E10696. [PMID: 29162697 DOI: 10.1073/pnas.1705682114] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Control of microtubule dynamics underlies several fundamental processes such as cell polarity, cell division, and cell motility. To gain insights into the mechanisms that control microtubule dynamics during cell motility, we investigated the interactome of the microtubule plus-end-binding protein end-binding 1 (EB1). Via molecular mapping and cross-linking mass spectrometry we identified and characterized a large complex associating a specific isoform of myomegalin termed "SMYLE" (for short myomegalin-like EB1 binding protein), the PKA scaffolding protein AKAP9, and the pericentrosomal protein CDK5RAP2. SMYLE was associated through an evolutionarily conserved N-terminal domain with AKAP9, which in turn was anchored at the centrosome via CDK5RAP2. SMYLE connected the pericentrosomal complex to the microtubule-nucleating complex (γ-TuRC) via Galectin-3-binding protein. SMYLE associated with nascent centrosomal microtubules to promote microtubule assembly and acetylation. Disruption of SMYLE interaction with EB1 or AKAP9 prevented microtubule nucleation and their stabilization at the leading edge of migrating cells. In addition, SMYLE depletion led to defective astral microtubules and abnormal orientation of the mitotic spindle and triggered G1 cell-cycle arrest, which might be due to defective centrosome integrity. As a consequence, SMYLE loss of function had a profound impact on tumor cell motility and proliferation, suggesting that SMYLE might be an important player in tumor progression.
Collapse
|