1
|
Sébastien M, Paquette AL, Prowse ENP, Hendricks AG, Brouhard GJ. Doublecortin restricts neuronal branching by regulating tubulin polyglutamylation. Nat Commun 2025; 16:1749. [PMID: 39966472 PMCID: PMC11836384 DOI: 10.1038/s41467-025-56951-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Doublecortin is a neuronal microtubule-associated protein that regulates microtubule structure in neurons. Mutations in Doublecortin cause lissencephaly and subcortical band heterotopia by impairing neuronal migration. We use CRISPR/Cas9 to knock-out the Doublecortin gene in induced pluripotent stem cells and differentiate the cells into cortical neurons. DCX-KO neurons show reduced velocities of nuclear movements and an increased number of neurites early in neuronal development, consistent with previous findings. Neurite branching is regulated by a host of microtubule-associated proteins, as well as by microtubule polymerization dynamics. However, EB comet dynamics are unchanged in DCX-KO neurons. Rather, we observe a significant reduction in α-tubulin polyglutamylation in DCX-KO neurons. Polyglutamylation levels and neuronal branching are rescued by expression of Doublecortin or of TTLL11, an α-tubulin glutamylase. Using U2OS cells as an orthogonal model system, we show that DCX and TTLL11 act synergistically to promote polyglutamylation. We propose that Doublecortin acts as a positive regulator of α-tubulin polyglutamylation and restricts neurite branching. Our results indicate an unexpected role for Doublecortin in the homeostasis of the tubulin code.
Collapse
Affiliation(s)
- Muriel Sébastien
- Department of Biology, McGill University, Montréal, QC, Canada
- Department of Bioengineering, McGill University, Montréal, QC, Canada
| | | | - Emily N P Prowse
- Department of Bioengineering, McGill University, Montréal, QC, Canada
| | - Adam G Hendricks
- Department of Bioengineering, McGill University, Montréal, QC, Canada
| | - Gary J Brouhard
- Department of Biology, McGill University, Montréal, QC, Canada.
| |
Collapse
|
2
|
Su KC, Radul E, Maier NK, Tsang MJ, Goul C, Moodie B, Marescal O, Keys HR, Cheeseman IM. Functional genetics reveals modulators of antimicrotubule drug sensitivity. J Cell Biol 2025; 224:e202403065. [PMID: 39570287 PMCID: PMC11590752 DOI: 10.1083/jcb.202403065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/04/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
Microtubules play essential roles in diverse cellular processes and are important pharmacological targets for treating human disease. Here, we sought to identify cellular factors that modulate the sensitivity of cells to antimicrotubule drugs. We conducted a genome-wide CRISPR/Cas9-based functional genetics screen in human cells treated with the microtubule-destabilizing drug nocodazole or the microtubule-stabilizing drug paclitaxel. We further conducted a focused secondary screen to test drug sensitivity for ∼1,400 gene targets across two distinct human cell lines and to additionally test sensitivity to the KIF11 inhibitor, STLC. These screens defined gene targets whose loss enhances or suppresses sensitivity to antimicrotubule drugs. In addition to gene targets whose loss sensitized cells to multiple compounds, we observed cases of differential sensitivity to specific compounds and differing requirements between cell lines. Our downstream molecular analysis further revealed additional roles for established microtubule-associated proteins and identified new players in microtubule function.
Collapse
Affiliation(s)
- Kuan-Chung Su
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Elena Radul
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Nolan K. Maier
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mary-Jane Tsang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Claire Goul
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Océane Marescal
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Heather R. Keys
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Iain M. Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
3
|
Onofre TS, Zhou Q, Li Z. The microtubule-severing enzyme spastin regulates spindle dynamics to promote chromosome segregation in Trypanosoma brucei. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.03.631140. [PMID: 39803587 PMCID: PMC11722300 DOI: 10.1101/2025.01.03.631140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Microtubule-severing enzymes play essential roles in regulating diverse cellular processes, including mitosis and cytokinesis, by modulating microtubule dynamics. In the early branching protozoan parasite Trypanosoma brucei, microtubule-severing enzymes are involved in cytokinesis and flagellum length control during different life cycle stages, but none of them have been found to regulate mitosis in any life cycle form. Here, we report the biochemical and functional characterization of the microtubule-severing enzyme spastin in the procyclic form of T. brucei. We demonstrate that spastin catalyzes microtubule severing in vitro and ectopic overexpression of spastin disrupts spindle microtubules in vivo in trypanosome cells, leading to defective chromosome segregation. Knockdown of spastin impairs spindle integrity and disrupts chromosome alignment in metaphase and chromosome segregation in anaphase. We further show that the function of spastin requires the catalytic AAA-ATPase domain, the microtubule-binding domain, and the microtubule interacting and trafficking domain, and that the association of spastin with spindle depends on the microtubule-binding domain. Together, these results uncover an essential role for spastin in chromosome segregation by regulating spindle dynamics in this unicellular eukaryote.
Collapse
Affiliation(s)
- Thiago Souza Onofre
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Qing Zhou
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Ziyin Li
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
4
|
Yasuda T, Nakajima N, Ogi T, Yanaka T, Tanaka I, Gotoh T, Kagawa W, Sugasawa K, Tajima K. Heavy water inhibits DNA double-strand break repairs and disturbs cellular transcription, presumably via quantum-level mechanisms of kinetic isotope effects on hydrolytic enzyme reactions. PLoS One 2024; 19:e0309689. [PMID: 39361575 PMCID: PMC11449287 DOI: 10.1371/journal.pone.0309689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/16/2024] [Indexed: 10/05/2024] Open
Abstract
Heavy water, containing the heavy hydrogen isotope, is toxic to cells, although the underlying mechanism remains incompletely understood. In addition, certain enzymatic proton transfer reactions exhibit kinetic isotope effects attributed to hydrogen isotopes and their temperature dependencies, indicative of quantum tunneling phenomena. However, the correlation between the biological effects of heavy water and the kinetic isotope effects mediated by hydrogen isotopes remains elusive. In this study, we elucidated the kinetic isotope effects arising from hydrogen isotopes of water and their temperature dependencies in vitro, focusing on deacetylation, DNA cleavage, and protein cleavage, which are crucial enzymatic reactions mediated by hydrolysis. Intriguingly, the intracellular isotope effects of heavy water, related to the in vitro kinetic isotope effects, significantly impeded multiple DNA double-strand break repair mechanisms crucial for cell survival. Additionally, heavy water exposure enhanced histone acetylation and associated transcriptional activation in cells, consistent with the in vitro kinetic isotope effects observed in histone deacetylation reactions. Moreover, as observed for the in vitro kinetic isotope effects, the cytotoxic effect on cell proliferation induced by heavy water exhibited temperature-dependency. These findings reveal the substantial impact of heavy water-induced isotope effects on cellular functions governed by hydrolytic enzymatic reactions, potentially mediated by quantum-level mechanisms underlying kinetic isotope effects.
Collapse
Affiliation(s)
- Takeshi Yasuda
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Nakako Nakajima
- QST Hospital, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Tomoko Yanaka
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Izumi Tanaka
- Institute for Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takaya Gotoh
- Department of Health Science, Daito Bunka University, Saitama, Japan
| | - Wataru Kagawa
- Department of Interdisciplinary Science and Engineering, Program in Chemistry and Life Science, School of Science and Engineering, Meisei University, Tokyo, Japan
| | - Kaoru Sugasawa
- Biosignal Research Center, and Graduate School of Science, Kobe University, Kobe, Japan
| | - Katsushi Tajima
- Department of Hematology, Yamagata Prefectural Central Hospital, Yamagata, Japan
| |
Collapse
|
5
|
Xie P. Modeling study of kinesin-13 MCAK microtubule depolymerase. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:339-354. [PMID: 39093405 DOI: 10.1007/s00249-024-01718-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Mitotic centromere-associated kinesin (MCAK) motor protein is a typical member of the kinesin-13 family, which can depolymerize microtubules from both plus and minus ends. A critical issue for the MCAK motor is how it performs the depolymerase activity. To address the issue, the pathway of the MCAK motor moving on microtubules and depolymerizing the microtubules is presented here. On the basis of the pathway, the dynamics of both the wild-type and mutant MCAK motors is studied theoretically, which include the full-length MCAK, the full-length MCAK with mutations in the α4-helix of the motor domain, the mutant full-length MCAK with a neutralized neck, the monomeric MCAK and the mutant monomeric MCAK with a neutralized neck. The studies show that a single dimeric MCAK motor can depolymerize microtubules in a processive manner, with either one tubulin or two tubulins being removed per times. The theoretical results are in agreement with the available experimental data. Moreover, predicted results are provided.
Collapse
Affiliation(s)
- Ping Xie
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
6
|
Zoroddu S, Sanna L, Bordoni V, Weidong L, Gadau SD, Carta A, Kelvin DJ, Bagella L. Identification of 3-Aryl-1-benzotriazole-1-yl-acrylonitrile as a Microtubule-Targeting Agent (MTA) in Solid Tumors. Int J Mol Sci 2024; 25:5704. [PMID: 38891892 PMCID: PMC11172098 DOI: 10.3390/ijms25115704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/06/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Recently, a compound derived from recent scientific advances named 34 has emerged as the focus of this research, the aim of which is to explore its potential impact on solid tumor cell lines. Using a combination of bioinformatics and biological assays, this study conducted an in-depth investigation of the effects of 34. The results of this study have substantial implications for cancer research and treatment. 34 has shown remarkable efficacy in inhibiting the growth of several cancer cell lines, including those representing prostate carcinoma (PC3) and cervical carcinoma (HeLa). The high sensitivity of these cells, indicated by low IC50 values, underscores its potential as a promising chemotherapeutic agent. In addition, 34 has revealed the ability to induce cell cycle arrest, particularly in the G2/M phase, a phenomenon with critical implications for tumor initiation and growth. By interfering with DNA replication in cancer cells, 34 has shown the capacity to trigger cell death, offering a new avenue for cancer treatment. In addition, computational analyses have identified key genes affected by 34 treatment, suggesting potential therapeutic targets. These genes are involved in critical biological processes, including cell cycle regulation, DNA replication and microtubule dynamics, all of which are central to cancer development and progression. In conclusion, this study highlights the different mechanisms of 34 that inhibit cancer cell growth and alter the cell cycle. These promising results suggest the potential for more effective and less toxic anticancer therapies. Further in vivo validation and exploration of combination therapies are critical to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Stefano Zoroddu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (L.S.); (V.B.)
| | - Luca Sanna
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (L.S.); (V.B.)
| | - Valentina Bordoni
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (L.S.); (V.B.)
| | - Lyu Weidong
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou 515031, China; (L.W.); (D.J.K.)
| | | | - Antonio Carta
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | - David J. Kelvin
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou 515031, China; (L.W.); (D.J.K.)
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Luigi Bagella
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (L.S.); (V.B.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Centre for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
7
|
Xie S, Yang Y, Jin Z, Liu X, Zhang S, Su N, Liu J, Li C, Zhang D, Gao L, Yang Z. Mouse KL2 is a unique MTSE involved in chromosome-based spindle organization and regulated by multiple kinases during female meiosis. J Biomed Res 2024; 38:1-15. [PMID: 38808565 PMCID: PMC11461529 DOI: 10.7555/jbr.37.20230290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 05/30/2024] Open
Abstract
Microtubule-severing enzymes (MTSEs) play important roles in mitosis and meiosis of the primitive organisms. However, no studies have assessed their roles in mammalian meiosis of females, whose abnormality accounts for over 80% of the cases of gamete-originated human reproductive disease. In the current study, we reported that katanin-like 2 (KL2) was the only MTSE concentrating at chromosomes. Furthermore, the knockdown of KL2 significantly reduced chromosome-based increase in the microtubule (MT) polymer, increased aberrant kinetochore-MT (K-MT) attachment, delayed meiosis, and severely affected normal fertility. Importantly, we demonstrated that the inhibition of aurora B, a key kinase for correcting aberrant K-MT attachment, eliminated KL2 from chromosomes completely. KL2 also interacted with phosphorylated eukaryotic elongation factor-2 kinase; they competed for chromosome binding. We also observed that the phosphorylated KL2 was localized at spindle poles, and that KL2 phosphorylation was regulated by extracellular signal-regulated kinase 1/2. In summary, our study reveals a novel function of MTSEs in mammalian female meiosis and demonstrates that multiple kinases coordinate to regulate the levels of KL2 at chromosomes.
Collapse
Affiliation(s)
- Shiya Xie
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Central Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Department of Gynaecology and Obstetrics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yanjie Yang
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Central Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Department of Gynaecology and Obstetrics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Zhen Jin
- Center for Reproductive Medicine, Department of Gynecology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaocong Liu
- Laboratory Department of Shihezi People's Hospital, Shihezi, Xinjiang 832099, China
| | - Shuping Zhang
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ning Su
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiaqi Liu
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Congrong Li
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Dong Zhang
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Gynaecology and Obstetrics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Leilei Gao
- Center for Reproductive Medicine, Department of Gynecology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Zhixia Yang
- Central Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| |
Collapse
|
8
|
Łomzik M, Błauż A, Tchoń D, Makal A, Rychlik B, Plażuk D. Development of Half-Sandwich Ru, Os, Rh, and Ir Complexes Bearing the Pyridine-2-ylmethanimine Bidentate Ligand Derived from 7-Chloroquinazolin-4(3H)-one with Enhanced Antiproliferative Activity. ACS OMEGA 2024; 9:18224-18237. [PMID: 38680348 PMCID: PMC11044151 DOI: 10.1021/acsomega.3c10482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024]
Abstract
Kinesin spindle protein (KSP) inhibitors are one of the most promising anticancer agents developed in recent years. Herein, we report the synthesis of ispinesib-core pyridine derivative conjugates, which are potent KSP inhibitors, with half-sandwich complexes of ruthenium, osmium, rhodium, and iridium. Conjugation of 7-chloroquinazolin-4(3H)-one with the pyridine-2-ylmethylimine group and the organometallic moiety resulted in up to a 36-fold increased cytotoxicity with IC50 values in the micromolar and nanomolar range also toward drug-resistant cells. All studied conjugates increased the percentage of cells in the G2/M phase, simultaneously decreasing the number of cells in the G1/G0 phase, suggesting mitotic arrest. Additionally, ruthenium derivatives were able to generate reactive oxygen species (ROS); however, no significant influence of the organometallic moiety on KSP inhibition was observed, which suggests that conjugation of a KSP inhibitor with the organometallic moiety modulates its mechanism of action.
Collapse
Affiliation(s)
- Michał Łomzik
- Faculty
of Chemistry, Department of Organic Chemistry, University of Lodz, ul. Tamka 12, 91-403 Łódź, Poland
| | - Andrzej Błauż
- Faculty
of Biology and Environmental Protection, Department of Oncobiology
and Epigenetics, Cytometry Lab, University
of Lodz, ul. Pomorska
141/143, 90-236 Łódź, Poland
| | - Daniel Tchoń
- Laboratory
for Structural and Biochemical Research (LBSBio), Biological and Chemical
Research Centre, Department of Chemistry, University of Warsaw, ul. Zwirki i Wigury 101, 02-089 Warszawa, Poland
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Anna Makal
- Laboratory
for Structural and Biochemical Research (LBSBio), Biological and Chemical
Research Centre, Department of Chemistry, University of Warsaw, ul. Zwirki i Wigury 101, 02-089 Warszawa, Poland
| | - Błażej Rychlik
- Faculty
of Biology and Environmental Protection, Department of Oncobiology
and Epigenetics, Cytometry Lab, University
of Lodz, ul. Pomorska
141/143, 90-236 Łódź, Poland
| | - Damian Plażuk
- Faculty
of Chemistry, Department of Organic Chemistry, University of Lodz, ul. Tamka 12, 91-403 Łódź, Poland
| |
Collapse
|
9
|
Su KC, Radul E, Maier NK, Tsang MJ, Goul C, Moodie B, Keys HR, Cheeseman IM. Functional genetics reveals modulators of anti-microtubule drug sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584469. [PMID: 38559203 PMCID: PMC10979949 DOI: 10.1101/2024.03.12.584469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Microtubules play essential roles in diverse cellular processes and are important pharmacological targets for treating human disease. Here, we sought to identify cellular factors that modulate the sensitivity of cells to anti-microtubule drugs. We conducted a genome-wide CRISPR/Cas9-based functional genetics screen in human cells treated with the microtubule-destabilizing drug nocodazole or the microtubule-stabilizing drug taxol. We further conducted a focused secondary screen to test drug sensitivity for ~1400 gene targets across two distinct human cell lines and to additionally test sensitivity to the Kif11-inhibitor, STLC. These screens defined gene targets whose loss enhances or suppresses sensitivity to anti-microtubule drugs. In addition to gene targets whose loss sensitized cells to multiple compounds, we observed cases of differential sensitivity to specific compounds and differing requirements between cell lines. Our downstream molecular analysis further revealed additional roles for established microtubule-associated proteins and identified new players in microtubule function.
Collapse
Affiliation(s)
- Kuan-Chung Su
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142
- These authors contributed equally
| | - Elena Radul
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142
- These authors contributed equally
- Present address: Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Nolan K Maier
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- These authors contributed equally
- Present address: Department of Microbiology, Harvard Medical School, Boston, MA
| | - Mary-Jane Tsang
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142
- These authors contributed equally
- Present address: Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Claire Goul
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Present address: Department of Molecular and Cellular Biology, UC Berkeley, Berkeley, CA
| | - Brittania Moodie
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142
| | - Heather R. Keys
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| |
Collapse
|
10
|
Zheng R, Xu FX, Zhou L, Xu J, Shen Y, Hao K, Wang XT, Deng J. Ablation of KIF2C in Purkinje cells impairs metabotropic glutamate receptor trafficking and motor coordination in male mice. J Physiol 2023; 601:3905-3920. [PMID: 37431690 DOI: 10.1113/jp284214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/27/2023] [Indexed: 07/12/2023] Open
Abstract
Kinesin family member 2C (KIF2C)/mitotic centromere-associated kinesin (MCAK), is thought to be oncogenic as it is involved in tumour progression and metastasis. Moreover, it also plays a part in neurodegenerative conditions like Alzheimer's disease and psychiatric disorders such as suicidal schizophrenia. Our previous study conducted on mice demonstrated that KIF2C is widely distributed in various regions of the brain, and is localized in synaptic spines. Additionally, it regulates microtubule dynamic properties through its own microtubule depolymerization activity, thereby affecting AMPA receptor transport and cognitive behaviour in mice. In this study, we show that KIF2C regulates the transport of mGlu1 receptors in Purkinje cells by binding to Rab8. KIF2C deficiency in Purkinje cells results in abnormal gait, reduced balance ability and motor incoordination in male mice. These data suggest that KIF2C is essential for maintaining normal transport and synaptic function of mGlu1 and motor coordination in mice. KEY POINTS: KIF2C is localized in synaptic spines of hippocampus neurons, and regulates excitatory transmission, synaptic plasticity and cognitive behaviour. KIF2C is extensively expressed in the cerebellum, and we investigated its functions in development and synaptic transmission of cerebellar Purkinje cells. KIF2C deficiency in Purkinje cells alters the expression of metabotropic glutamate receptor 1 (mGlu1) and the AMPA receptor GluA2 subunit at Purkinje cell synapses, and changes excitatory synaptic transmission, but not inhibitory transmission. KIF2C regulates the transport of mGlu1 receptors in Purkinje cells by binding to Rab8. KIF2C deficiency in Purkinje cells affects motor coordination, but not social behaviour in male mice.
Collapse
Affiliation(s)
- Rui Zheng
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| | - Fang-Xiao Xu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Lin Zhou
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Junyu Xu
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Ying Shen
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ke Hao
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xin-Tai Wang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, China
| | - Junjie Deng
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| |
Collapse
|
11
|
Al-Hiyasat A, Tuna Y, Kuo YW, Howard J. Herding of proteins by the ends of shrinking polymers. Phys Rev E 2023; 107:L042601. [PMID: 37198784 PMCID: PMC11550390 DOI: 10.1103/physreve.107.l042601] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/17/2023] [Indexed: 05/19/2023]
Abstract
The control of biopolymer length is mediated by proteins that localize to polymer ends and regulate polymerization dynamics. Several mechanisms have been proposed to achieve end localization. Here, we propose a novel mechanism by which a protein that binds to a shrinking polymer and slows its shrinkage will be spontaneously enriched at the shrinking end through a "herding" effect. We formalize this process using both lattice-gas and continuum descriptions, and we present experimental evidence that the microtubule regulator spastin employs this mechanism. Our findings extend to more general problems involving diffusion within shrinking domains.
Collapse
Affiliation(s)
- Amer Al-Hiyasat
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, USA
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Yazgan Tuna
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, USA
| | - Yin-Wei Kuo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, USA
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
12
|
Chew WX, Henkin G, Nédélec F, Surrey T. Effects of microtubule length and crowding on active microtubule network organization. iScience 2023; 26:106063. [PMID: 36852161 PMCID: PMC9958361 DOI: 10.1016/j.isci.2023.106063] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/12/2022] [Accepted: 01/20/2023] [Indexed: 01/28/2023] Open
Abstract
Active filament networks can organize into various dynamic architectures driven by cross-linking motors. Densities and kinetic properties of motors and microtubules have been shown previously to determine active microtubule network self-organization, but the effects of other control parameters are less understood. Using computer simulations, we study here how microtubule lengths and crowding effects determine active network architecture and dynamics. We find that attractive interactions mimicking crowding effects or long microtubules both promote the formation of extensile nematic networks instead of asters. When microtubules are very long and the network is highly connected, a new isotropically motile network state resembling a "gliding mesh" is predicted. Using in vitro reconstitutions, we confirm the existence of this gliding mesh experimentally. These results provide a better understanding of how active microtubule network organization can be controlled, with implications for cell biology and active materials in general.
Collapse
Affiliation(s)
- Wei-Xiang Chew
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain
| | - Gil Henkin
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain
| | - François Nédélec
- Sainsbury Laboratory, University of Cambridge, 47 Bateman Street, Cambridge CB2 1LR, UK,Corresponding author
| | - Thomas Surrey
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain,ICREA, Passeig de Lluis Companys 23, 08010 Barcelona, Spain,Corresponding author
| |
Collapse
|
13
|
Schaer J, Andreu-Carbó M, Kruse K, Aumeier C. The effect of motor-induced shaft dynamics on microtubule stability and length. Biophys J 2023; 122:346-359. [PMID: 36502273 PMCID: PMC9892620 DOI: 10.1016/j.bpj.2022.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Control of microtubule abundance, stability, and length is crucial to regulate intracellular transport as well as cell polarity and division. How microtubule stability depends on tubulin addition or removal at the dynamic ends is well studied. However, microtubule rescue, the event when a microtubule switches from shrinking to growing, occurs at tubulin exchange sites along the shaft. Molecular motors have recently been shown to promote such exchanges. Using a stochastic theoretical description, we study how microtubule stability and length depend on motor-induced tubulin exchange and thus rescue. Our theoretical description matches our in vitro experiments on microtubule dynamics in the presence of kinesin-1 molecular motors. Although the overall dynamics of a population of microtubules can be captured by an effective rescue rate, by assigning rescue to exchange sites, we reveal that the dynamics of individual microtubules within the population differ dramatically. Furthermore, we study in detail a transition from bounded to unbounded microtubule growth. Our results provide novel insights into how molecular motors imprint information of microtubule stability on the microtubule network.
Collapse
Affiliation(s)
- Joël Schaer
- Department of Biochemistry, University of Geneva, Geneva, Switzerland; Department of Theoretical Physics, University of Geneva, Geneva, Switzerland
| | | | - Karsten Kruse
- Department of Biochemistry, University of Geneva, Geneva, Switzerland; Department of Theoretical Physics, University of Geneva, Geneva, Switzerland; National Center for Competence in Research Chemical Biology, University of Geneva, Geneva, Switzerland.
| | - Charlotte Aumeier
- Department of Biochemistry, University of Geneva, Geneva, Switzerland; National Center for Competence in Research Chemical Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
14
|
Xie P. A model for the catalytic activity of microtubule polymerases. Cytoskeleton (Hoboken) 2023; 80:7-20. [PMID: 36305831 DOI: 10.1002/cm.21734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/04/2022] [Accepted: 10/22/2022] [Indexed: 11/07/2022]
Abstract
A XMAP215/Stu2/Alp14 polymerase can catalyze processively the tubulin addition to the microtubule (MT) plus end. In this work, a model is proposed for the underlying molecular mechanism of the polymerase activity, where the polymerase can not only catalyze processively the tubulin addition to but also promote the tubulin removal from the MT plus end. Based on the model the dynamics of both the wild-type and mutant polymerases is studied theoretically, explaining consistently and well various available experimental data. To further test the model, predicted results are provided.
Collapse
Affiliation(s)
- Ping Xie
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Science, Beijing, China
| |
Collapse
|
15
|
Striebel M, Brauns F, Frey E. Length Regulation Drives Self-Organization in Filament-Motor Mixtures. PHYSICAL REVIEW LETTERS 2022; 129:238102. [PMID: 36563230 DOI: 10.1103/physrevlett.129.238102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/10/2022] [Indexed: 06/17/2023]
Abstract
Cytoskeletal networks form complex intracellular structures. Here we investigate a minimal model for filament-motor mixtures in which motors act as depolymerases and thereby regulate filament length. Combining agent-based simulations and hydrodynamic equations, we show that resource-limited length regulation drives the formation of filament clusters despite the absence of mechanical interactions between filaments. Even though the orientation of individual remains fixed, collective filament orientation emerges in the clusters, aligned orthogonal to their interfaces.
Collapse
Affiliation(s)
- Moritz Striebel
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of Physics, Ludwig-Maximilians-Universität München, Theresienstrasse 37, D-80333 Munich, Germany
| | - Fridtjof Brauns
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of Physics, Ludwig-Maximilians-Universität München, Theresienstrasse 37, D-80333 Munich, Germany
| | - Erwin Frey
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of Physics, Ludwig-Maximilians-Universität München, Theresienstrasse 37, D-80333 Munich, Germany
- Max Planck School Matter to Life, Hofgartenstraße 8, D-80539 Munich, Germany
| |
Collapse
|
16
|
Microtubule-severing protein Fidgetin-like 1 promotes spindle organization during meiosis of mouse oocytes. ZYGOTE 2022; 30:872-881. [PMID: 36148793 DOI: 10.1017/s0967199422000417] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Microtubule-severing proteins (MTSPs) play important roles in mitosis and interphase. However, to the best of our knowledge, no previous studies have evaluated the role of MTSPs in female meiosis in mammals. It was found that FIGNL1, a member of MTSPs, was predominantly expressed in mouse oocytes and distributed at the spindle poles during meiosis in the present study. FIGNL1 was co-localized and interacted with γ-tubulin, an important component of the microtubule tissue centre (MTOC). Fignl1 knockdown by specific small interfering RNA caused spindle defects characterized by an abnormal length:width ratio and decreased microtubule density, which consequently led to aberrant chromosome arrangement, oocyte maturation and fertilization obstacles. In conclusion, the present results suggested that FIGNL1 may be an essential factor in oocyte maturation by influencing the meiosis process via the formation of spindles.
Collapse
|
17
|
Costa AC, Sousa MM. The Role of Spastin in Axon Biology. Front Cell Dev Biol 2022; 10:934522. [PMID: 35865632 PMCID: PMC9294387 DOI: 10.3389/fcell.2022.934522] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/07/2022] [Indexed: 12/05/2022] Open
Abstract
Neurons are highly polarized cells with elaborate shapes that allow them to perform their function. In neurons, microtubule organization—length, density, and dynamics—are essential for the establishment of polarity, growth, and transport. A mounting body of evidence shows that modulation of the microtubule cytoskeleton by microtubule-associated proteins fine tunes key aspects of neuronal cell biology. In this respect, microtubule severing enzymes—spastin, katanin and fidgetin—a group of microtubule-associated proteins that bind to and generate internal breaks in the microtubule lattice, are emerging as key modulators of the microtubule cytoskeleton in different model systems. In this review, we provide an integrative view on the latest research demonstrating the key role of spastin in neurons, specifically in the context of axonal cell biology. We focus on the function of spastin in the regulation of microtubule organization, and axonal transport, that underlie its importance in the intricate control of axon growth, branching and regeneration.
Collapse
Affiliation(s)
- Ana Catarina Costa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
- Graduate Program in Molecular and Cell Biology, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- *Correspondence: Ana Catarina Costa, ; Monica Mendes Sousa,
| | - Monica Mendes Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
- *Correspondence: Ana Catarina Costa, ; Monica Mendes Sousa,
| |
Collapse
|
18
|
Size regulation of multiple organelles competing for a limiting subunit pool. PLoS Comput Biol 2022; 18:e1010253. [PMID: 35714135 PMCID: PMC9246132 DOI: 10.1371/journal.pcbi.1010253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/30/2022] [Accepted: 05/26/2022] [Indexed: 11/20/2022] Open
Abstract
How cells regulate the size of intracellular structures and organelles is a longstanding question. Recent experiments suggest that size control of intracellular structures is achieved through the depletion of a limiting subunit pool in the cytoplasm. While the limiting pool model ensures organelle-to-cell size scaling, it does not provide a mechanism for robust size control of multiple co-existing structures. Here we develop a generalized theory for size-dependent growth of intracellular structures to demonstrate that robust size control of multiple intracellular structures, competing for a limiting subunit pool, is achieved via a negative feedback between the growth rate and the size of the individual structure. This design principle captures size maintenance of a wide variety of subcellular structures, from cytoskeletal filaments to three-dimensional organelles. We identify the feedback motifs for structure size regulation based on known molecular processes, and compare our theory to existing models of size regulation in biological assemblies. Furthermore, we show that positive feedback between structure size and growth rate can lead to bistable size distribution and spontaneous size selection. Organelle size control is essential for the proper physiological functioning of eukaryotic cells, but the underlying mechanisms of size regulation remain poorly understood. By developing a general theory for organelle size control, we show that robust size control of intracellular structures and organelles is achieved via a negative feedback between individual organelle size and their net growth rates. This design principle not only describes size maintenance of single organelles, but also ensures size stability of multiple co-existing organelles that are built from a limiting pool of subunits. Our results delineate the role of limiting pool as a size scaling mechanism rather than a size control mechanism, supporting the idea that negative feedback control of organelle size via depletion of a limiting subunit pool is not sufficient to maintain the size of multiple competing organelles. In the case of positive feedback between organelle size and growth rate, our model reproduces phenomena such as bistability in organelle size distribution and spontaneous emergence of cell polarity.
Collapse
|
19
|
Banerjee DS, Banerjee S. Emergence and maintenance of variable-length actin filaments in a limiting pool of building blocks. Biophys J 2022; 121:2436-2448. [PMID: 35598045 DOI: 10.1016/j.bpj.2022.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 04/10/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Actin is one of the key structural components of the eukaryotic cytoskeleton that regulates cellular architecture and mechanical properties. Dynamic regulation of actin filament length and organization is essential for the control of many physiological processes including cell adhesion, motility and division. While previous studies have mostly focused on the mechanisms controlling the mean length of individual actin filaments, it remains poorly understood how distinct actin filament populations in cells maintain different lengths using the same set of molecular building blocks. Here we develop a theoretical model for the length regulation of multiple actin filaments by nucleation and growth rate modulation by actin binding proteins in a limiting pool of monomers. We first show that spontaneous nucleation of actin filaments naturally leads to heterogeneities in filament length distribution. We then investigate the effects of filament growth inhibition by capping proteins and growth promotion by formin proteins on filament length distribution. We find that filament length heterogeneity can be increased by growth inhibition, whereas growth promoters do not significantly affect length heterogeneity. Interestingly, a competition between filament growth inhibitors and growth promoters can give rise to bimodal filament length distribution as well as a highly heterogeneous length distribution with large statistical dispersion. We quantitatively predict how heterogeneity in actin filament length can be modulated by tuning F-actin nucleation and growth rates in order to create distinct filament subpopulations with different lengths. SIGNIFICANCE: Actin filaments organize into different functional network architectures within eukaryotic cells. To maintain distinct actin network architectures, it is essential to regulate the lengths of actin filaments. While the mechanisms controlling the lengths of individual actin filaments have been extensively studied, the regulation of length heterogeneity in actin filament populations is not well understood. Here we show that the modulation of actin filament growth and nucleation rates by actin binding proteins can regulate actin length distribution and create distinct sub-populations with different lengths. In particular, by tuning concentrations of formin, profilin and capping proteins, various aspects of actin filament length distribution can be controlled. Insights gained from our results may have significant implications for the regulation of actin filament length heterogeneity and architecture within a cell.
Collapse
Affiliation(s)
- Deb Sankar Banerjee
- Carnegie Mellon University, Department of Physics, Pittsburgh, PA 15213, USA
| | - Shiladitya Banerjee
- Carnegie Mellon University, Department of Physics, Pittsburgh, PA 15213, USA
| |
Collapse
|
20
|
Zheng R, Du Y, Wang X, Liao T, Zhang Z, Wang N, Li X, Shen Y, Shi L, Luo J, Xia J, Wang Z, Xu J. KIF2C regulates synaptic plasticity and cognition in mice through dynamic microtubule depolymerization. eLife 2022; 11:e72483. [PMID: 35138249 PMCID: PMC8828051 DOI: 10.7554/elife.72483] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Dynamic microtubules play a critical role in cell structure and function. In nervous system, microtubules are the major route for cargo protein trafficking and they specially extend into and out of synapses to regulate synaptic development and plasticity. However, the detailed depolymerization mechanism that regulates dynamic microtubules in synapses and dendrites is still unclear. In this study, we find that KIF2C, a dynamic microtubule depolymerization protein without known function in the nervous system, plays a pivotal role in the structural and functional plasticity of synapses and regulates cognitive function in mice. Through its microtubule depolymerization capability, KIF2C regulates microtubule dynamics in dendrites, and regulates microtubule invasion of spines in neurons in a neuronal activity-dependent manner. Using RNAi knockdown and conditional knockout approaches, we showed that KIF2C regulates spine morphology and synaptic membrane expression of AMPA receptors. Moreover, KIF2C deficiency leads to impaired excitatory transmission, long-term potentiation, and altered cognitive behaviors in mice. Collectively, our study explores a novel function of KIF2C in the nervous system and provides an important regulatory mechanism on how activity-dependent microtubule dynamic regulates synaptic plasticity and cognition behaviors.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Yonglan Du
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Xintai Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Tailin Liao
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Zhe Zhang
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Na Wang
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Xiumao Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Ying Shen
- Department of Physiology and Department of Neurology of First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Lei Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan UniversityGuanzhouChina
| | - Jianhong Luo
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Jun Xia
- Division of Life Science and The Brain and Intelligence Research Institute, The Hong Kong University of Science and TechnologyHong KongChina
| | - Ziyi Wang
- Innovative Institute of Basic Medical Sciences of Zhejiang University (Yuhang)HangzhouChina
| | - Junyu Xu
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| |
Collapse
|
21
|
Atomic force microscopy reveals distinct protofilament-scale structural dynamics in depolymerizing microtubule arrays. Proc Natl Acad Sci U S A 2022; 119:2115708119. [PMID: 35101922 PMCID: PMC8812519 DOI: 10.1073/pnas.2115708119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 12/01/2022] Open
Abstract
One cannot help but marvel at the precise organization of microtubule polymers in cellular structures such as the axoneme and the spindle. However, our understanding of the biochemical mechanisms that sculpt these arrays comes largely from in vitro experiments with a small number (one or two) of microtubules. This is somewhat akin to studying the architecture of multilane highways by studying one-lane streets. Here, we directly visualize depolymerizing microtubule arrays at individual microtubule and protofilament resolution using atomic force microscopy. Our results reveal differences in microtubule depolymerase activity and provide insights into how these differences in enzymatic activity on the nanometer scale can result in the differential remodeling of multimicrotubule arrays on the micron-length scale. The dynamic reorganization of microtubule-based cellular structures, such as the spindle and the axoneme, fundamentally depends on the dynamics of individual polymers within multimicrotubule arrays. A major class of enzymes implicated in both the complete demolition and fine size control of microtubule-based arrays are depolymerizing kinesins. How different depolymerases differently remodel microtubule arrays is poorly understood. A major technical challenge in addressing this question is that existing optical or electron-microscopy methods lack the spatial-temporal resolution to observe the dynamics of individual microtubules within larger arrays. Here, we use atomic force microscopy (AFM) to image depolymerizing arrays at single-microtubule and protofilament resolution. We discover previously unseen modes of microtubule array destabilization by conserved depolymerases. We find that the kinesin-13 MCAK mediates asynchronous protofilament depolymerization and lattice-defect propagation, whereas the kinesin-8 Kip3p promotes synchronous protofilament depolymerization. Unexpectedly, MCAK can depolymerize the highly stable axonemal doublets, but Kip3p cannot. We propose that distinct protofilament-level activities underlie the functional dichotomy of depolymerases, resulting in either large-scale destabilization or length regulation of microtubule arrays. Our work establishes AFM as a powerful strategy to visualize microtubule dynamics within arrays and reveals how nanometer-scale substrate specificity leads to differential remodeling of micron-scale cytoskeletal structures.
Collapse
|
22
|
McAlear TS, Bechstedt S. The mitotic spindle protein CKAP2 potently increases formation and stability of microtubules. eLife 2022; 11:72202. [PMID: 35029146 PMCID: PMC8798059 DOI: 10.7554/elife.72202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Cells increase microtubule dynamics to make large rearrangements to their microtubule cytoskeleton during cell division. Changes in microtubule dynamics are essential for the formation and function of the mitotic spindle, and misregulation can lead to aneuploidy and cancer. Using in vitro reconstitution assays we show that the mitotic spindle protein Cytoskeleton-Associated Protein 2 (CKAP2) has a strong effect on nucleation of microtubules by lowering the critical tubulin concentration 100-fold. CKAP2 increases the apparent rate constant ka of microtubule growth by 50-fold and increases microtubule growth rates. In addition, CKAP2 strongly suppresses catastrophes. Our results identify CKAP2 as the most potent microtubule growth factor to date. These finding help explain CKAP2's role as an important spindle protein, proliferation marker, and oncogene.
Collapse
Affiliation(s)
- Thomas S McAlear
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Susanne Bechstedt
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| |
Collapse
|
23
|
Motor usage imprints microtubule stability along the shaft. Dev Cell 2021; 57:5-18.e8. [PMID: 34883065 DOI: 10.1016/j.devcel.2021.11.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/27/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022]
Abstract
Tubulin dimers assemble into dynamic microtubules, which are used by molecular motors as tracks for intracellular transport. Organization and dynamics of the microtubule network are commonly thought to be regulated at the polymer ends, where tubulin dimers can be added or removed. Here, we show that molecular motors running on microtubules cause exchange of dimers along the shaft in vitro and in cells. These sites of dimer exchange act as rescue sites where depolymerizing microtubules stop shrinking and start re-growing. Consequently, the average length of microtubules increases depending on how frequently they are used as motor tracks. An increase of motor activity densifies the cellular microtubule network and enhances cell polarity. Running motors leave marks in the shaft, serving as traces of microtubule usage to organize the polarity landscape of the cell.
Collapse
|
24
|
Chen B, Xie X, Lan F, Liu W. Identification of prognostic markers by weighted gene co-expression network analysis in non-small cell lung cancer. Bioengineered 2021; 12:4924-4935. [PMID: 34369264 PMCID: PMC8806742 DOI: 10.1080/21655979.2021.1960764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the fatal tumors and is associated with a poor prognosis. Cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) was used to quantify the proportions of 22 types of immune cells. Weighted gene co-expression network analysis (WGCNA) was established from the GSE37745 data, and key modules correlating most with CD8+ T cell infiltration were determined. Genes that manifested a high module connectivity in the key module were identified as hub genes. Three bioinformatics online databases were used to evaluate hub gene expression levels in tumor and normal tissues. Finally, survival analysis was conducted for these hub genes. In this study, we chose four hub genes (AURKB, CDC20, TPX2 and KIF2C) based on the comprehensive bioinformatics analyses. All hub genes were overexpressed in tumor tissue, and high expression of AURKB, CDC20, TPX2, and KIF2C correlated with the poor prognosis of these patients. In vitro experiments confirmed that CDC20 knockdown inhibited cell proliferation and growth. The above results indicated that AURKB, CDC20, TPX2, and KIF2C are potential CD8+ T cell infiltration-related biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Binglin Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaowei Xie
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Feifeng Lan
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenqi Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
25
|
Mani N, Wijeratne SS, Subramanian R. Micron-scale geometrical features of microtubules as regulators of microtubule organization. eLife 2021; 10:e63880. [PMID: 34114950 PMCID: PMC8195601 DOI: 10.7554/elife.63880] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 06/02/2021] [Indexed: 12/20/2022] Open
Abstract
The organization of micron-sized, multi-microtubule arrays from individual microtubules is essential for diverse cellular functions. The microtubule polymer is largely viewed as a passive building block during the organization process. An exception is the 'tubulin code' where alterations to tubulin at the amino acid level can influence the activity of microtubule-associated proteins. Recent studies reveal that micron-scale geometrical features of individual microtubules and polymer networks, such as microtubule length, overlap length, contact angle, and lattice defects, can also regulate the activity of microtubule-associated proteins and modulate polymer dynamics. We discuss how the interplay between such geometrical properties of the microtubule lattice and the activity of associated proteins direct multiple aspects of array organization, from microtubule nucleation and coalignment to specification of array dimensions and remodeling of dynamic networks. The mechanisms reviewed here highlight micron-sized features of microtubules as critical parameters to be routinely investigated in the study of microtubule self-organization.
Collapse
Affiliation(s)
- Nandini Mani
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Sithara S Wijeratne
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
26
|
Hsu NY, Pathak N, Chen YT, Hsu YC, Yang JM. Pharmacophore anchor models of ATAT1 to discover potential inhibitors and lead optimization. Comput Biol Chem 2021; 93:107513. [PMID: 34052673 DOI: 10.1016/j.compbiolchem.2021.107513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 11/30/2022]
Abstract
Post-translation modification of microtubules is associated with many diseases like cancer. Alpha Tubulin Acetyltransferase 1 (ATAT1) is a major enzyme that acetylates 'Lys-40' in alpha-tubulin on the luminal side of microtubules and is a drug target that lacks inhibitors. Here, we developed pharmacophore anchor models of ATAT1 which were constructed statistically using thousands of docked compounds, for drug design and investigating binding mechanisms. Our models infer the compound moiety preferences with the physico-chemical properties for the ATAT1 binding site. The results from the pharmacophore anchor models show the three main sub-pockets, including S1 acetyl site, S2 adenine site, and S3 diphosphate site with anchors, where conserved moieties interact with respective sub-pocket residues in each site and help in guiding inhibitor discovery. We validated these key anchors by analyzing 162 homologous protein sequences (>99 species) and over 10 structures with various bound ligands and mutations. Our results were consistent with previous works also providing new interesting insights. Our models applied in virtual screening predicted several ATAT1 potential inhibitors. We believe that our model is useful for future inhibitor discovery and for guiding lead optimization.
Collapse
Affiliation(s)
- Nung-Yu Hsu
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 30050, Taiwan; Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 30050, Taiwan
| | - Nikhil Pathak
- TIGP-Bioinformatics, Institute of Information Science, Academia Sinica, Taipei, 115, Taiwan; Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yun-Ti Chen
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 30050, Taiwan; Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 30050, Taiwan
| | - Yen-Chao Hsu
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 30050, Taiwan; Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 30050, Taiwan
| | - Jinn-Moon Yang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 30050, Taiwan; Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 30050, Taiwan.
| |
Collapse
|
27
|
Barisic M, Rajendraprasad G, Steblyanko Y. The metaphase spindle at steady state - Mechanism and functions of microtubule poleward flux. Semin Cell Dev Biol 2021; 117:99-117. [PMID: 34053864 DOI: 10.1016/j.semcdb.2021.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 11/24/2022]
Abstract
The mitotic spindle is a bipolar cellular structure, built from tubulin polymers, called microtubules, and interacting proteins. This macromolecular machine orchestrates chromosome segregation, thereby ensuring accurate distribution of genetic material into the two daughter cells during cell division. Powered by GTP hydrolysis upon tubulin polymerization, the microtubule ends exhibit a metastable behavior known as the dynamic instability, during which they stochastically switch between the growth and shrinkage phases. In the context of the mitotic spindle, dynamic instability is furthermore regulated by microtubule-associated proteins and motor proteins, which enables the spindle to undergo profound changes during mitosis. This highly dynamic behavior is essential for chromosome capture and congression in prometaphase, as well as for chromosome alignment to the spindle equator in metaphase and their segregation in anaphase. In this review we focus on the mechanisms underlying microtubule dynamics and sliding and their importance for the maintenance of shape, structure and dynamics of the metaphase spindle. We discuss how these spindle properties are related to the phenomenon of microtubule poleward flux, highlighting its highly cooperative molecular basis and role in keeping the metaphase spindle at a steady state.
Collapse
Affiliation(s)
- Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center (DCRC), Strandboulevarden 49, 2100 Copenhagen, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| | - Girish Rajendraprasad
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center (DCRC), Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Yulia Steblyanko
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center (DCRC), Strandboulevarden 49, 2100 Copenhagen, Denmark
| |
Collapse
|
28
|
Spastin interacts with CRMP5 to promote spindle organization in mouse oocytes by severing microtubules. ZYGOTE 2021; 30:80-91. [PMID: 34034836 DOI: 10.1017/s0967199421000344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microtubule-severing protein (MTSP) is critical for the survival of both mitotic and postmitotic cells. However, the study of MTSP during meiosis of mammalian oocytes has not been reported. We found that spastin, a member of the MTSP family, was highly expressed in oocytes and aggregated in spindle microtubules. After knocking down spastin by specific siRNA, the spindle microtubule density of meiotic oocytes decreased significantly. When the oocytes were cultured in vitro, the oocytes lacking spastin showed an obvious maturation disorder. Considering the microtubule-severing activity of spastin, we speculate that spastin on spindles may increase the number of microtubule broken ends by severing the microtubules, therefore playing a nucleating role, promoting spindle assembly and ensuring normal meiosis. In addition, we found the colocalization and interaction of collapsin response mediator protein 5 (CRMP5) and spastin in oocytes. CRMP5 can provide structural support and promote microtubule aggregation, creating transportation routes, and can interact with spastin in the microtubule activity of nerve cells (30). Knocking down CRMP5 may lead to spindle abnormalities and developmental disorders in oocytes. Overexpression of spastin may reverse the abnormal phenotype caused by the deletion of CRMP5. In summary, our data support a model in which the interaction between spastin and CRMP5 promotes the assembly of spindle microtubules in oocytes by controlling microtubule dynamics, therefore ensuring normal meiosis.
Collapse
|
29
|
Albahde MAH, Abdrakhimov B, Li GQ, Zhou X, Zhou D, Xu H, Qian H, Wang W. The Role of Microtubules in Pancreatic Cancer: Therapeutic Progress. Front Oncol 2021; 11:640863. [PMID: 34094924 PMCID: PMC8176010 DOI: 10.3389/fonc.2021.640863] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer has an extremely low prognosis, which is attributable to its high aggressiveness, invasiveness, late diagnosis, and lack of effective therapies. Among all the drugs joining the fight against this type of cancer, microtubule-targeting agents are considered to be the most promising. They inhibit cancer cells although through different mechanisms such as blocking cell division, apoptosis induction, etc. Hereby, we review the functions of microtubule cytoskeletal proteins in tumor cells and comprehensively examine the effects of microtubule-targeting agents on pancreatic carcinoma.
Collapse
Affiliation(s)
- Mugahed Abdullah Hasan Albahde
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
| | - Bulat Abdrakhimov
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guo-Qi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Xiaohu Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Dongkai Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Hao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Huixiao Qian
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
30
|
Minagawa M, Shirato M, Toya M, Sato M. Dual Impact of a Benzimidazole Resistant β-Tubulin on Microtubule Behavior in Fission Yeast. Cells 2021; 10:1042. [PMID: 33925026 PMCID: PMC8145593 DOI: 10.3390/cells10051042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 11/17/2022] Open
Abstract
The cytoskeleton microtubule consists of polymerized αβ-tubulin dimers and plays essential roles in many cellular events. Reagents that inhibit microtubule behaviors have been developed as antifungal, antiparasitic, and anticancer drugs. Benzimidazole compounds, including thiabendazole (TBZ), carbendazim (MBC), and nocodazole, are prevailing microtubule poisons that target β-tubulin and inhibit microtubule polymerization. The molecular basis, however, as to how the drug acts on β-tubulin remains controversial. Here, we characterize the S. pombe β-tubulin mutant nda3-TB101, which was previously isolated as a mutant resistance to benzimidazole. The mutation site tyrosine at position 50 is located in the interface of two lateral β-tubulin proteins and at the gate of a putative binging pocket for benzimidazole. Our observation revealed two properties of the mutant tubulin. First, the dynamics of cellular microtubules comprising the mutant β-tubulin were stabilized in the absence of benzimidazole. Second, the mutant protein reduced the affinity to benzimidazole in vitro. We therefore conclude that the mutant β-tubulin Nda3-TB101 exerts a dual effect on microtubule behaviors: the mutant β-tubulin stabilizes microtubules and is insensitive to benzimidazole drugs. This notion fine-tunes the current elusive molecular model regarding binding of benzimidazole to β-tubulin.
Collapse
Affiliation(s)
- Mamika Minagawa
- Laboratory of Cytoskeletal Logistics, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan; (M.M.); (M.S.); (M.T.)
| | - Minamo Shirato
- Laboratory of Cytoskeletal Logistics, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan; (M.M.); (M.S.); (M.T.)
| | - Mika Toya
- Laboratory of Cytoskeletal Logistics, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan; (M.M.); (M.S.); (M.T.)
- Faculty of Science and Engineering, Global Center for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Masamitsu Sato
- Laboratory of Cytoskeletal Logistics, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan; (M.M.); (M.S.); (M.T.)
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Institute for Medical-Oriented Structural Biology, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan
| |
Collapse
|
31
|
Habicht J, Mooneyham A, Hoshino A, Shetty M, Zhang X, Emmings E, Yang Q, Coombes C, Gardner MK, Bazzaro M. UNC-45A breaks the microtubule lattice independently of its effects on non-muscle myosin II. J Cell Sci 2021; 134:jcs.248815. [PMID: 33262310 DOI: 10.1242/jcs.248815] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022] Open
Abstract
In invertebrates, UNC-45 regulates myosin stability and functions. Vertebrates have two distinct isoforms of the protein: UNC-45B, expressed in muscle cells only, and UNC-45A, expressed in all cells and implicated in regulating both non-muscle myosin II (NMII)- and microtubule (MT)-associated functions. Here, we show that, in vitro and in human and rat cells, UNC-45A binds to the MT lattice, leading to MT bending, breakage and depolymerization. Furthermore, we show that UNC-45A destabilizes MTs independent of its C-terminal NMII-binding domain and even in the presence of the NMII inhibitor blebbistatin. These findings identified UNC-45A as a novel type of MT-severing protein with a dual non-mutually exclusive role in regulating NMII activity and MT stability. Because many human diseases, from cancer to neurodegenerative diseases, are caused by or associated with deregulation of MT stability, our findings have profound implications in the biology of MTs, as well as the biology of human diseases and possible therapeutic implications for their treatment.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Juri Habicht
- Bradenburg Medical School - Theodor Fontane, Neuruppin 16816, Germany.,Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ashley Mooneyham
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Asumi Hoshino
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mihir Shetty
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Xiaonan Zhang
- Bradenburg Medical School - Theodor Fontane, Neuruppin 16816, Germany.,Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Edith Emmings
- Bradenburg Medical School - Theodor Fontane, Neuruppin 16816, Germany
| | - Qing Yang
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Courtney Coombes
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Melissa K Gardner
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Martina Bazzaro
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
32
|
Kuo YW, Howard J. Cutting, Amplifying, and Aligning Microtubules with Severing Enzymes. Trends Cell Biol 2020; 31:50-61. [PMID: 33183955 PMCID: PMC7749064 DOI: 10.1016/j.tcb.2020.10.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023]
Abstract
Microtubule-severing enzymes - katanin, spastin, fidgetin - are related AAA-ATPases that cut microtubules into shorter filaments. These proteins, also called severases, are involved in a wide range of cellular processes including cell division, neuronal development, and tissue morphogenesis. Paradoxically, severases can amplify the microtubule cytoskeleton and not just destroy it. Recent work on spastin and katanin has partially resolved this paradox by showing that these enzymes are strong promoters of microtubule growth. Here, we review recent structural and biophysical advances in understanding the molecular mechanisms of severing and growth promotion that provide insight into how severing enzymes shape microtubule networks.
Collapse
Affiliation(s)
- Yin-Wei Kuo
- Department of Chemistry, Yale University, New Haven, CT 06511, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
33
|
Meng F, Zhang L, Ren Y, Ma Q. Transcriptome analysis reveals key signature genes involved in the oncogenesis of lung cancer. Cancer Biomark 2020; 29:475-482. [PMID: 32831194 DOI: 10.3233/cbm-200110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Previous studies have suggested potential signature genes for lung cancer, however, due to factors such as sequencing platform, control, data selection and filtration conditions, the results of lung cancer-related gene expression analysis are quite different. Here, we performed a meta-analysis on existing lung cancer gene expression results to identify Meta-signature genes without noise. In this study, functional enrichment, protein-protein interaction network, the DAVID, String, TfactS, and transcription factor binding were performed based on the gene expression profiles of lung adenocarcinoma and non-small cell lung cancer deposited in the GEO database. As a result, a total of 574 differentially expressed genes (DEGs) affecting the pathogenesis of lung cancer were identified (207 up-regulated expression and 367 down-regulated expression in lung cancer tissues). A total of 5,093 interactions existed among the 507 (88.3%) proteins, and 10 Meta-signatures were identified: AURKA, CCNB1, KIF11, CCNA2, TOP2A, CENPF, KIF2C, TPX2, HMMR, and MAD2L1. The potential biological functions of Meta-signature DEGs were revealed. In summary, this study identified key genes involved in the process of lung cancer. Our results would help the developing of novel biomarkers for lung cancer.
Collapse
|
34
|
Forkosh E, Kenig A, Ilan Y. Introducing variability in targeting the microtubules: Review of current mechanisms and future directions in colchicine therapy. Pharmacol Res Perspect 2020; 8:e00616. [PMID: 32608157 PMCID: PMC7327382 DOI: 10.1002/prp2.616] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022] Open
Abstract
Microtubules (MTs) are highly dynamic polymers that constitute the cellular cytoskeleton and play a role in multiple cellular functions. Variability characterizes biological systems and is considered a part of the normal function of cells and organs. Variability contributes to cell plasticity and is a mechanism for overcoming errors in cellular level assembly and function, and potentially the whole organ level. Dynamic instability is a feature of biological variability that characterizes the function of MTs. The dynamic behavior of MTs constitutes the basis for multiple biological processes that contribute to cellular plasticity and the timing of cell signaling. Colchicine is a MT-modifying drug that exerts anti-inflammatory and anti-cancer effects. This review discusses some of the functions of colchicine and presents a platform for introducing variability while targeting MTs in intestinal cells, the microbiome, the gut, and the systemic immune system. This platform can be used for implementing novel therapies, improving response to chronic MT-based therapies, overcoming drug resistance, exerting gut-based systemic immune responses, and generating patient-tailored dynamic therapeutic regimens.
Collapse
Affiliation(s)
- Esther Forkosh
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| | - Ariel Kenig
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| | - Yaron Ilan
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| |
Collapse
|
35
|
Thawani A, Rale MJ, Coudray N, Bhabha G, Stone HA, Shaevitz JW, Petry S. The transition state and regulation of γ-TuRC-mediated microtubule nucleation revealed by single molecule microscopy. eLife 2020; 9:e54253. [PMID: 32538784 PMCID: PMC7338055 DOI: 10.7554/elife.54253] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 06/15/2020] [Indexed: 12/31/2022] Open
Abstract
Determining how microtubules (MTs) are nucleated is essential for understanding how the cytoskeleton assembles. While the MT nucleator, γ-tubulin ring complex (γ-TuRC) has been identified, precisely how γ-TuRC nucleates a MT remains poorly understood. Here, we developed a single molecule assay to directly visualize nucleation of a MT from purified Xenopus laevis γ-TuRC. We reveal a high γ-/αβ-tubulin affinity, which facilitates assembly of a MT from γ-TuRC. Whereas spontaneous nucleation requires assembly of 8 αβ-tubulins, nucleation from γ-TuRC occurs efficiently with a cooperativity of 4 αβ-tubulin dimers. This is distinct from pre-assembled MT seeds, where a single dimer is sufficient to initiate growth. A computational model predicts our kinetic measurements and reveals the rate-limiting transition where laterally associated αβ-tubulins drive γ-TuRC into a closed conformation. NME7, TPX2, and the putative activation domain of CDK5RAP2 h γ-TuRC-mediated nucleation, while XMAP215 drastically increases the nucleation efficiency by strengthening the longitudinal γ-/αβ-tubulin interaction.
Collapse
Affiliation(s)
- Akanksha Thawani
- Department of Chemical and Biological Engineering, Princeton UniversityPrincetonUnited States
| | - Michael J Rale
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Nicolas Coudray
- Department of Cell Biology, New York University School of MedicineNew YorkUnited States
| | - Gira Bhabha
- Department of Cell Biology, New York University School of MedicineNew YorkUnited States
| | - Howard A Stone
- Department of Mechanical and Aerospace Engineering, Princeton UniversityPrincetonUnited States
| | - Joshua W Shaevitz
- Lewis-Sigler Institute for Integrative GenomicsPrincetonUnited States
- Department of Physics, Princeton UniversityPrincetonUnited States
| | - Sabine Petry
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| |
Collapse
|
36
|
Stiff T, Echegaray-Iturra FR, Pink HJ, Herbert A, Reyes-Aldasoro CC, Hochegger H. Prophase-Specific Perinuclear Actin Coordinates Centrosome Separation and Positioning to Ensure Accurate Chromosome Segregation. Cell Rep 2020; 31:107681. [PMID: 32460023 PMCID: PMC7262599 DOI: 10.1016/j.celrep.2020.107681] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 02/11/2020] [Accepted: 05/01/2020] [Indexed: 12/30/2022] Open
Abstract
Centrosome separation in late G2/ early prophase requires precise spatial coordination that is determined by a balance of forces promoting and antagonizing separation. The major effector of centrosome separation is the kinesin Eg5. However, the identity and regulation of Eg5-antagonizing forces is less well characterized. By manipulating candidate components, we find that centrosome separation is reversible and that separated centrosomes congress toward a central position underneath the flat nucleus. This positioning mechanism requires microtubule polymerization, as well as actin polymerization. We identify perinuclear actin structures that form in late G2/early prophase and interact with microtubules emanating from the centrosomes. Disrupting these structures by breaking the interactions of the linker of nucleoskeleton and cytoskeleton (LINC) complex with perinuclear actin filaments abrogates this centrosome positioning mechanism and causes an increase in subsequent chromosome segregation errors. Our results demonstrate how geometrical cues from the cell nucleus coordinate the orientation of the emanating spindle poles before nuclear envelope breakdown.
Collapse
Affiliation(s)
- Tom Stiff
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Fabio R Echegaray-Iturra
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Harry J Pink
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Alex Herbert
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | | | - Helfrid Hochegger
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK.
| |
Collapse
|
37
|
Lin Y, Wei YL, She ZY. Kinesin-8 motors: regulation of microtubule dynamics and chromosome movements. Chromosoma 2020; 129:99-110. [PMID: 32417983 DOI: 10.1007/s00412-020-00736-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/01/2023]
Abstract
Microtubules are essential for intracellular transport, cell motility, spindle assembly, and chromosome segregation during cell division. Microtubule dynamics regulate the proper spindle organization and thus contribute to chromosome congression and segregation. Accumulating studies suggest that kinesin-8 motors are emerging regulators of microtubule dynamics and organizations. In this review, we provide an overview of the studies focused on kinesin-8 motors in cell division. We discuss the structures and molecular kinetics of kinesin-8 motors. We highlight the essential roles and mechanisms of kinesin-8 in the regulation of microtubule dynamics and spindle organization. We also shed light on the functions of kinesin-8 motors in chromosome movement and the spindle assembly checkpoint during the cell cycle.
Collapse
Affiliation(s)
- Yang Lin
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.,Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Ya-Lan Wei
- Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350011, Fujian, China.,Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China. .,Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
38
|
Tempes A, Weslawski J, Brzozowska A, Jaworski J. Role of dynein-dynactin complex, kinesins, motor adaptors, and their phosphorylation in dendritogenesis. J Neurochem 2020; 155:10-28. [PMID: 32196676 DOI: 10.1111/jnc.15010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/24/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
One of the characteristic features of different classes of neurons that is vital for their proper functioning within neuronal networks is the shape of their dendritic arbors. To properly develop dendritic trees, neurons need to accurately control the intracellular transport of various cellular cargo (e.g., mRNA, proteins, and organelles). Microtubules and motor proteins (e.g., dynein and kinesins) that move along microtubule tracks play an essential role in cargo sorting and transport to the most distal ends of neurons. Equally important are motor adaptors, which may affect motor activity and specify cargo that is transported by the motor. Such transport undergoes very dynamic fine-tuning in response to changes in the extracellular environment and synaptic transmission. Such regulation is achieved by the phosphorylation of motors, motor adaptors, and cargo, among other mechanisms. This review focuses on the contribution of the dynein-dynactin complex, kinesins, their adaptors, and the phosphorylation of these proteins in the formation of dendritic trees by maturing neurons. We primarily review the effects of the motor activity of these proteins in dendrites on dendritogenesis. We also discuss less anticipated mechanisms that contribute to dendrite growth, such as dynein-driven axonal transport and non-motor functions of kinesins.
Collapse
Affiliation(s)
- Aleksandra Tempes
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jan Weslawski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Agnieszka Brzozowska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jacek Jaworski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| |
Collapse
|
39
|
Tang Q, Rui M, Bu S, Wang Y, Chew LY, Yu F. A microtubule polymerase is required for microtubule orientation and dendrite pruning in Drosophila. EMBO J 2020; 39:e103549. [PMID: 32267553 DOI: 10.15252/embj.2019103549] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 01/12/2023] Open
Abstract
Drosophila class IV ddaC neurons selectively prune all larval dendrites to refine the nervous system during metamorphosis. During dendrite pruning, severing of proximal dendrites is preceded by local microtubule (MT) disassembly. Here, we identify an unexpected role of Mini spindles (Msps), a conserved MT polymerase, in governing dendrite pruning. Msps associates with another MT-associated protein TACC, and both stabilize each other in ddaC neurons. Moreover, Msps and TACC are required to orient minus-end-out MTs in dendrites. We further show that the functions of msps in dendritic MT orientation and dendrite pruning are antagonized by the kinesin-13 MT depolymerase Klp10A. Excessive MT depolymerization, which is induced by pharmacological treatment and katanin overexpression, also perturbs dendritic MT orientation and dendrite pruning, phenocopying msps mutants. Thus, we demonstrate that the MT polymerase Msps is required to form dendritic minus-end-out MTs and thereby promotes dendrite pruning in Drosophila sensory neurons.
Collapse
Affiliation(s)
- Quan Tang
- Temasek Life Sciences Laboratory, Singapore City, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore City, Singapore
| | - Menglong Rui
- Temasek Life Sciences Laboratory, Singapore City, Singapore
| | - Shufeng Bu
- Temasek Life Sciences Laboratory, Singapore City, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore City, Singapore
| | - Yan Wang
- Temasek Life Sciences Laboratory, Singapore City, Singapore
| | - Liang Yuh Chew
- Temasek Life Sciences Laboratory, Singapore City, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore City, Singapore
| | - Fengwei Yu
- Temasek Life Sciences Laboratory, Singapore City, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore City, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, Singapore City, Singapore.,Neuroscience and Behavioral Disorder Program, Duke-NUS Graduate Medical School Singapore, Singapore City, Singapore
| |
Collapse
|
40
|
Zhang G, Shen S, Yu Y, Yue X, Hu W, Li S. Kinesin family member 2C aggravates the progression of hepatocellular carcinoma and interacts with competing endogenous RNA. J Cell Biochem 2020; 121:4419-4430. [DOI: 10.1002/jcb.29665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Guo‐Pei Zhang
- Department of Liver Surgery the First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Shun‐Li Shen
- Department of Liver Surgery the First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Yang Yu
- Department of Liver Surgery the First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Xiao Yue
- Department of Liver Surgery the First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Wen‐Jie Hu
- Department of Liver Surgery the First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Shao‐Qiang Li
- Department of Liver Surgery the First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| |
Collapse
|
41
|
Vukušić K, Buđa R, Tolić IM. Force-generating mechanisms of anaphase in human cells. J Cell Sci 2019; 132:132/18/jcs231985. [DOI: 10.1242/jcs.231985] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
ABSTRACT
What forces drive chromosome segregation remains one of the most challenging questions in cell division. Even though the duration of anaphase is short, it is of utmost importance for genome fidelity that no mistakes are made. Seminal studies in model organisms have revealed different mechanisms operating during chromosome segregation in anaphase, but the translation of these mechanisms to human cells is not straightforward. Recent work has shown that kinetochore fiber depolymerization during anaphase A is largely motor independent, whereas spindle elongation during anaphase B is coupled to sliding of interpolar microtubules in human cells. In this Review, we discuss the current knowledge on the mechanisms of force generation by kinetochore, interpolar and astral microtubules. By combining results from numerous studies, we propose a comprehensive picture of the role of individual force-producing and -regulating proteins. Finally, by linking key concepts of anaphase to most recent data, we summarize the contribution of all proposed mechanisms to chromosome segregation and argue that sliding of interpolar microtubules and depolymerization at the kinetochore are the main drivers of chromosome segregation during early anaphase in human cells.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Renata Buđa
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Iva M. Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| |
Collapse
|
42
|
Chen X, Widmer LA, Stangier MM, Steinmetz MO, Stelling J, Barral Y. Remote control of microtubule plus-end dynamics and function from the minus-end. eLife 2019; 8:48627. [PMID: 31490122 PMCID: PMC6754230 DOI: 10.7554/elife.48627] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022] Open
Abstract
In eukaryotes, the organization and function of the microtubule cytoskeleton depend on the allocation of different roles to individual microtubules. For example, many asymmetrically dividing cells differentially specify microtubule behavior at old and new centrosomes. Here we show that yeast spindle pole bodies (SPBs, yeast centrosomes) differentially control the plus-end dynamics and cargoes of their astral microtubules, remotely from the minus-end. The old SPB recruits the kinesin motor protein Kip2, which then translocates to the plus-end of the emanating microtubules, promotes their extension and delivers dynein into the bud. Kip2 recruitment at the SPB depends on Bub2 and Bfa1, and phosphorylation of cytoplasmic Kip2 prevents random lattice binding. Releasing Kip2 of its control by SPBs equalizes its distribution, the length of microtubules and dynein distribution between the mother cell and its bud. These observations reveal that microtubule organizing centers use minus to plus-end directed remote control to individualize microtubule function.
Collapse
Affiliation(s)
- Xiuzhen Chen
- Institute of Biochemistry, ETH Zürich, Zurich, Switzerland
| | - Lukas A Widmer
- Department of Biosystems Science and Engineering, ETH Zürich, SIB Swiss Institute of Bioinformatics, Basel, Switzerland.,Systems Biology PhD Program, Life Science Zurich Graduate School, Zurich, Switzerland
| | - Marcel M Stangier
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland.,Biozentrum, University of Basel, Basel, Switzerland
| | - Jörg Stelling
- Department of Biosystems Science and Engineering, ETH Zürich, SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Yves Barral
- Institute of Biochemistry, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
43
|
Hemmat M, Castle BT, Sachs JN, Odde DJ. Multiscale Computational Modeling of Tubulin-Tubulin Lateral Interaction. Biophys J 2019; 117:1234-1249. [PMID: 31493861 DOI: 10.1016/j.bpj.2019.08.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/26/2019] [Accepted: 08/13/2019] [Indexed: 10/26/2022] Open
Abstract
Microtubules are multistranded polymers in eukaryotic cells that support key cellular functions such as chromosome segregation, motor-based cargo transport, and maintenance of cell polarity. Microtubules self-assemble via "dynamic instability," in which the dynamic plus ends switch stochastically between alternating phases of polymerization and depolymerization. A key question in the field is what are the atomistic origins of this switching, i.e., what is different between the GTP- and GDP-tubulin states that enables microtubule growth and shortening, respectively? More generally, a major challenge in biology is how to connect theoretical frameworks across length- and timescales, from atoms to cellular behavior. In this study, we describe a multiscale model by linking atomistic molecular dynamics (MD), molecular Brownian dynamics (BD), and cellular-level thermokinetic modeling of microtubules. Here, we investigated the underlying interaction energy when tubulin dimers associate laterally by performing all-atom MD simulations. We found that the lateral potential energy is not significantly different among three nucleotide states of tubulin, GTP, GDP, and GMPCPP and is estimated to be ≅ -11 kBT. Furthermore, using MD potential energy in our BD simulations of tubulin dimers confirms that the lateral bond is weak on its own, with a mean lifetime of ∼0.1 μs, implying that the longitudinal bond is required for microtubule assembly. We conclude that nucleotide-dependent lateral-bond strength is not the key mediator microtubule dynamic instability, implying that GTP acts elsewhere to exert its stabilizing influence on microtubule polymer. Furthermore, the estimated lateral-bond strength (ΔGlat0≅ -5 kBT) is well-aligned with earlier estimates based on thermokinetic modeling and light microscopy measurements. Thus, we have computationally connected atomistic-level structural information, obtained by cryo-electron microscopy, to cellular-scale microtubule assembly dynamics using a combination of MD, BD, and thermokinetic models to bridge from Ångstroms to micrometers and from femtoseconds to minutes.
Collapse
Affiliation(s)
- Mahya Hemmat
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Brian T Castle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Jonathan N Sachs
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
44
|
Ilan-Ber T, Ilan Y. The role of microtubules in the immune system and as potential targets for gut-based immunotherapy. Mol Immunol 2019; 111:73-82. [DOI: 10.1016/j.molimm.2019.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 12/18/2022]
|
45
|
Verma AK, Sharma N, Gupta AK. Cooperative motor action to regulate microtubule length dynamics. Phys Rev E 2019; 99:032411. [PMID: 30999491 DOI: 10.1103/physreve.99.032411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Indexed: 11/07/2022]
Abstract
Motivated by the recent experimental observations on motor induced cooperative mechanism controlling the length dynamics of microtubules (MTs), we examine how plus-end-targeted proteins of the kinesin family regulate MT polymerization and depolymerization routines. Here, we study a stochastic mathematical model capturing the unusual form of collective motor interaction on MT dynamics originating due to the molecular traffic near the MT tip. We provide an extensive analysis of the joint effect of motor impelled MT polymerization and complete depolymerization. The effect of the cooperative action is included by modifying the intrinsic depolymerization rate. We analyze the model within the framework of continuum mean-field theory and the resultant steady-state analytic solution is expressed in terms of Lambert W functions. Four distinct steady-state phases including a shock phase have been reported. The significant features of the shock including its position and height have been analyzed. Theoretical outcomes are supported by extensive Monte Carlo simulations. To explore the system alterations between the regime of growth and shrinkage phase, we consider kymographs of the microtubule along with the length distributions. Finally, we investigated the dependence of MT length kinetics both on modifying factor of depolymerization rate and motor concentration. The overall extensive study reveals that the flux of molecular traffic at the microtubule plus end initiates a cooperative mechanism, resulting in significant change in MT growth and shrinkage regime as also observed experimentally.
Collapse
Affiliation(s)
- Atul Kumar Verma
- Department of Mathematics, Indian Institute of Technology Ropar, Rupnagar-140001, Punjab, India
| | - Natasha Sharma
- Department of Mathematics, Indian Institute of Technology Ropar, Rupnagar-140001, Punjab, India
| | - Arvind Kumar Gupta
- Department of Mathematics, Indian Institute of Technology Ropar, Rupnagar-140001, Punjab, India
| |
Collapse
|
46
|
Pavlova GA, Razuvaeva AV, Popova JV, Andreyeva EN, Yarinich LA, Lebedev MO, Pellacani C, Bonaccorsi S, Somma MP, Gatti M, Pindyurin AV. The role of Patronin in Drosophila mitosis. BMC Mol Cell Biol 2019; 20:7. [PMID: 31284878 PMCID: PMC6469034 DOI: 10.1186/s12860-019-0189-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background The calmodulin-regulated spectrin-associated proteins (CAMSAPs) belong to a conserved protein family, which includes members that bind the polymerizing mcrotubule (MT) minus ends and remain associated with the MT lattice formed by minus end polymerization. Only one of the three mammalian CAMSAPs, CAMSAP1, localizes to the mitotic spindle but its function is unclear. In Drosophila, there is only one CAMSAP, named Patronin. Previous work has shown that Patronin stabilizes the minus ends of non-mitotic MTs and is required for proper spindle elongation. However, the precise role of Patronin in mitotic spindle assembly is poorly understood. Results Here we have explored the role of Patronin in Drosophila mitosis using S2 tissue culture cells as a model system. We show that Patronin associates with different types of MT bundles within the Drosophila mitotic spindle, and that it is required for their stability. Imaging of living cells expressing Patronin-GFP showed that Patronin displays a dynamic behavior. In prometaphase cells, Patronin accumulates on short segments of MT bundles located near the chromosomes. These Patronin “seeds” extend towards the cell poles and stop growing just before reaching the poles. Our data also suggest that Patronin localization is largely independent of proteins acting at the MT minus ends such as Asp and Klp10A. Conclusion Our results suggest a working hypothesis about the mitotic role of Patronin. We propose that Patronin binds the minus ends within MT bundles, including those generated from the walls of preexisting MTs via the augmin-mediated pathway. This would help maintaining MT association within the mitotic bundles, thereby stabilizing the spindle structure. Our data also raise the intriguing possibility that the minus ends of bundled MTs can undergo a limited polymerization. Electronic supplementary material The online version of this article (10.1186/s12860-019-0189-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gera A Pavlova
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Alyona V Razuvaeva
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.,Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Julia V Popova
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Evgeniya N Andreyeva
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Lyubov A Yarinich
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.,Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Mikhail O Lebedev
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.,Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Claudia Pellacani
- IBPM CNR and Department of Biology and Biotechnology, Sapienza University of Rome, 00185, Rome, Italy
| | - Silvia Bonaccorsi
- IBPM CNR and Department of Biology and Biotechnology, Sapienza University of Rome, 00185, Rome, Italy
| | - Maria Patrizia Somma
- IBPM CNR and Department of Biology and Biotechnology, Sapienza University of Rome, 00185, Rome, Italy
| | - Maurizio Gatti
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia. .,IBPM CNR and Department of Biology and Biotechnology, Sapienza University of Rome, 00185, Rome, Italy.
| | - Alexey V Pindyurin
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia. .,Novosibirsk State University, Novosibirsk, 630090, Russia.
| |
Collapse
|
47
|
Spastin is a dual-function enzyme that severs microtubules and promotes their regrowth to increase the number and mass of microtubules. Proc Natl Acad Sci U S A 2019; 116:5533-5541. [PMID: 30837315 PMCID: PMC6431158 DOI: 10.1073/pnas.1818824116] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The remodeling of the microtubule cytoskeleton underlies dynamic cellular processes, such as mitosis, ciliogenesis, and neuronal morphogenesis. An important class of microtubule remodelers comprises the severases-spastin, katanin, and fidgetin-which cut microtubules into shorter fragments. While severing activity might be expected to break down the microtubule cytoskeleton, inhibiting these enzymes in vivo actually decreases, rather increases, the number of microtubules, suggesting that severases have a nucleation-like activity. To resolve this paradox, we reconstituted Drosophila spastin in a dynamic microtubule assay and discovered that it is a dual-function enzyme. In addition to its ATP-dependent severing activity, spastin is an ATP-independent regulator of microtubule dynamics that slows shrinkage and increases rescue. We observed that spastin accumulates at shrinking ends; this increase in spastin concentration may underlie the increase in rescue frequency and the slowdown in shortening. The changes in microtubule dynamics promote microtubule regrowth so that severed microtubule fragments grow, leading to an increase in the number and mass of microtubules. A mathematical model shows that spastin's effect on microtubule dynamics is essential for this nucleation-like activity: spastin switches microtubules into a state where the net flux of tubulin onto each polymer is positive, leading to the observed exponential increase in microtubule mass. This increase in the microtubule mass accounts for spastin's in vivo phenotypes.
Collapse
|
48
|
Gao M, Berghaus M, Möbitz S, Schuabb V, Erwin N, Herzog M, Julius K, Sternemann C, Winter R. On the Origin of Microtubules' High-Pressure Sensitivity. Biophys J 2019. [PMID: 29539395 DOI: 10.1016/j.bpj.2018.01.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
For over 50 years, it has been known that the mitosis of eukaryotic cells is inhibited already at high hydrostatic pressure conditions of 30 MPa. This effect has been attributed to the disorganization of microtubules, the main component of the spindle apparatus. However, the structural details of the depolymerization and the origin of the pressure sensitivity have remained elusive. It has also been a puzzle how complex organisms could still successfully inhabit extreme high-pressure environments such as those encountered in the depth of oceans. We studied the pressure stability of microtubules at different structural levels and for distinct dynamic states using high-pressure Fourier-transform infrared spectroscopy and Synchrotron small-angle x-ray scattering. We show that microtubules are hardly stable under abyssal conditions, where pressures up to 100 MPa are reached. This high-pressure sensitivity can be mainly attributed to the internal voids and packing defects in the microtubules. In particular, we show that lateral and longitudinal contacts feature different pressure stabilities, and they define also the pressure stability of tubulin bundles. The intactness of both contact types is necessary for the functionality of microtubules in vivo. Despite being known to dynamically stabilize microtubules and prevent their depolymerization, we found that the anti-cancer drug taxol and the accessory protein MAP2c decrease the pressure stability of microtubule protofilaments. Moreover, we demonstrate that the cellular environment itself is a crowded place and accessory proteins can increase the pressure stability of microtubules and accelerate their otherwise highly pressure-sensitive de novo formation.
Collapse
Affiliation(s)
- Mimi Gao
- Physical Chemistry I - Biophysical Chemistry, Faculty of Chemistry and Chemical Biology
| | - Melanie Berghaus
- Physical Chemistry I - Biophysical Chemistry, Faculty of Chemistry and Chemical Biology
| | - Simone Möbitz
- Physical Chemistry I - Biophysical Chemistry, Faculty of Chemistry and Chemical Biology
| | - Vitor Schuabb
- Physical Chemistry I - Biophysical Chemistry, Faculty of Chemistry and Chemical Biology
| | - Nelli Erwin
- Physical Chemistry I - Biophysical Chemistry, Faculty of Chemistry and Chemical Biology
| | - Marius Herzog
- Physical Chemistry I - Biophysical Chemistry, Faculty of Chemistry and Chemical Biology
| | - Karin Julius
- Fakultät Physik/DELTA, Technische Universität Dortmund, Dortmund, Germany
| | | | - Roland Winter
- Physical Chemistry I - Biophysical Chemistry, Faculty of Chemistry and Chemical Biology.
| |
Collapse
|
49
|
Serikbaeva A, Tvorogova A, Kauanova S, Vorobjev IA. Analysis of Microtubule Dynamics Heterogeneity in Cell Culture. Methods Mol Biol 2019; 1745:181-204. [PMID: 29476470 DOI: 10.1007/978-1-4939-7680-5_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Microtubules (MTs) are dynamic components of the cytoskeleton playing an important role in a large number of cell functions. Individual MTs in living cells undergo stochastic switching between alternate states of growth, shortening and attenuated phase, a phenomenon known as tempered dynamic instability. Dynamic instability of MTs is usually analyzed by labeling MTs with +TIPs, namely, EB proteins. Tracking of +TIP trajectories allows analyzing MT growth in cells with a different density of MTs. Numerous labs now use +TIP to track growing MTs in a variety of cell cultures. However, heterogeneity of MT dynamics is usually underestimated, and rather small sampling for the description of dynamic instability parameters is often used. The strategy described in this chapter is the method for repetitive quantitative analysis of MT growth rate within the same cell that allows minimization of the variation in MT dynamics measurement. We show that variability in MT dynamics within a cell when using repeated measurements is significantly less than between different cells in the same chamber. This approach allows better estimation of the heterogeneity of cells' responses to different treatments. To compare the effects of different MT inhibitors, the protocol using normalized values for MT dynamics and repetitive measurements for each cell is employed. This chapter provides detailed methods for analysis of MT dynamics in tissue cultures. We describe protocols for imaging MT dynamics by fluorescent microscopy, contrast enhancement technique, and MT dynamics analysis using triple color-coded display based on sequential subtraction analysis.
Collapse
Affiliation(s)
- Anara Serikbaeva
- Department of Biology, School of Science and Technology, Nazarbayev University, Astana, Kazakhstan
- Department of Biology, School of Sciences and Technology, Nazarbayev University, Astana, Kazakhstan
| | - Anna Tvorogova
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Sholpan Kauanova
- School of Engineering, Nazarbayev University, Astana, Kazakhstan
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Ivan A Vorobjev
- Department of Biology, School of Sciences and Technology, Nazarbayev University, Astana, Kazakhstan.
| |
Collapse
|
50
|
Nozal V, Martinez A. Tau Tubulin Kinase 1 (TTBK1), a new player in the fight against neurodegenerative diseases. Eur J Med Chem 2019; 161:39-47. [DOI: 10.1016/j.ejmech.2018.10.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/09/2018] [Accepted: 10/12/2018] [Indexed: 10/28/2022]
|