1
|
Solomon AD, Gouttia OG, Wang L, Zhu S, Wang F, Li Y, Paydar M, Bessho T, Kwok BH, Peng A. γ-tubulin mediates DNA double-strand break repair. J Cell Sci 2025; 138:jcs262255. [PMID: 40135584 PMCID: PMC12050090 DOI: 10.1242/jcs.262255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 12/20/2024] [Indexed: 03/27/2025] Open
Abstract
Double-strand breaks (DSBs) in DNA pose a critical threat to genomic integrity, potentially leading to the onset and progression of various diseases, including cancer. Cellular responses to such lesions entail sophisticated repair mechanisms primarily mediated by non-homologous end joining (NHEJ) and homologous recombination (HR). Interestingly, the efficient recruitment of repair proteins and completion of DSB repair likely involve complex, inter-organelle communication and coordination of cellular components. In this study, we report a role of γ-tubulin in DSB repair. γ-tubulin is a major microtubule nucleation factor governing microtubule dynamics. We show that γ-tubulin is recruited to the site of DNA damage and is required for efficient DSB repair via both NHEJ and HR. Suppression of γ-tubulin impedes DNA repair and exacerbates DNA damage accumulation. Furthermore, γ-tubulin mediates the mobilization and formation of DNA damage foci, which serve as repair centers, thereby facilitating the recruitment of HR and NHEJ repair proteins on damaged chromatin. Finally, pharmacological inhibition of γ-tubulin enhances the cytotoxic effect of DNA-damaging agents, consistent with the DNA repair function of γ-tubulin, and underscoring the potential of its therapeutic intervention in cancer therapy.
Collapse
Affiliation(s)
- Abhishikt David Solomon
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Odjo G. Gouttia
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Wang
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Songli Zhu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Feifei Wang
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Yanqui Li
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Mohammadjavad Paydar
- Institute for Research in Immunology and Cancer (IRIC), Département de médecine, Université de Montréal, Montréal H3C 3J7, Canada
| | - Tadayoshi Bessho
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin H. Kwok
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Aimin Peng
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Lindström L, Zhou J, Villoutreix BO, Malycheva D, Otrocka M, Gustavsson A, Lundbäck T, Bliman D, Shameem MA, Straw M, Riesbeck K, Olsson R, Alvarado‐Kristensson M. Targeting TUBG1 in RB1-negative tumors. FASEB J 2025; 39:e70431. [PMID: 40019206 PMCID: PMC11869804 DOI: 10.1096/fj.202403180rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
The disruption of microtubule dynamics serves as a pivotal strategy for eliminating tumor cells, despite its accompanying toxicities affecting non-tumor cells. This study investigates the potential of selectively targeting γ-tubulin1 (TUBG1) as a therapeutic strategy in cancer treatment. By elucidating the TUBG1-E2F1-retinoblastoma protein (RB1) network, we introduce a novel compound, 4-(6-((3-Methoxyphenyl)amino)pyrimidin-4-yl)-N,N-dimethylbenzenamine, (L12). L12 treatment enhanced RB1 expression and selectively targeted cells with impaired RB1 signaling, while reduced E2F1 expression attenuated its cytotoxicity. Furthermore, L12-mediated cytotoxicity depends on an E2F1-mediated upregulation of procaspase 3 expression, highlighting the role of E2F1 in the apoptotic response. Unlike traditional tubulin-targeting agents, L12's specificity for tumor cells lies in its inhibitory effects on TUBG1, without affecting the second human isoform of TUBGs, TUBG2. Despite its interaction with specific kinases, the concentrations required for antitumor effects are 100-fold lower than those influencing kinase activities. Subsequent investigations underscore L12's reduced neuronal axonal toxicity compared to vincristine. Lastly, L12 demonstrates promising results in inhibiting tumor growth in xenografted small cell lung cancer models, demonstrating potential specificity toward tumor cells while minimizing adverse effects on healthy tissues. This research emphasizes the potential of TUBG1 inhibitors as a promising advancement in personalized chemotherapy approaches and their potential as a groundbreaking treatment for various cancers.
Collapse
Affiliation(s)
- Lisa Lindström
- Molecular Pathology, Department of Translational MedicineLund UniversityMalmöSweden
| | - Jingkai Zhou
- Molecular Pathology, Department of Translational MedicineLund UniversityMalmöSweden
| | | | - Darina Malycheva
- Molecular Pathology, Department of Translational MedicineLund UniversityMalmöSweden
| | - Magdalena Otrocka
- Chemical Biology Consortium Sweden, Science for Life LaboratoryKarolinska InstitutetSolnaSweden
| | - Anna‐Lena Gustavsson
- Chemical Biology Consortium Sweden, Science for Life LaboratoryKarolinska InstitutetSolnaSweden
| | - Thomas Lundbäck
- Chemical Biology Consortium Sweden, Science for Life LaboratoryKarolinska InstitutetSolnaSweden
| | - David Bliman
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
| | | | - Megan Straw
- Clinical Microbiology, Department of Translational MedicineLund UniversityMalmöSweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational MedicineLund UniversityMalmöSweden
| | - Roger Olsson
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
- Chemical Biology & Therapeutics, Department of Experimental Medical ScienceLund UniversityLundSweden
| | | |
Collapse
|
3
|
Gao Q, Hofer FW, Filbeck S, Vermeulen BJA, Würtz M, Neuner A, Kaplan C, Zezlina M, Sala C, Shin H, Gruss OJ, Schiebel E, Pfeffer S. Structural mechanisms for centrosomal recruitment and organization of the microtubule nucleator γ-TuRC. Nat Commun 2025; 16:2453. [PMID: 40074789 PMCID: PMC11903878 DOI: 10.1038/s41467-025-57729-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The γ-tubulin ring complex (γ-TuRC) acts as a structural template for microtubule formation at centrosomes, associating with two main compartments: the pericentriolar material and the centriole lumen. In the pericentriolar material, the γ-TuRC is involved in microtubule organization, while the function of the centriole lumenal pool remains unclear. The conformational landscape of the γ-TuRC, which is crucial for its activity, and its centrosomal anchoring mechanisms, which determine γ-TuRC activity and turnover, are not understood. Using cryo-electron tomography, we analyze γ-TuRCs in human cells and purified centrosomes. Pericentriolar γ-TuRCs simultaneously associate with the essential adapter NEDD1 and the microcephaly protein CDK5RAP2. NEDD1 forms a tetrameric structure at the γ-TuRC base through interactions with four GCP3/MZT1 modules and GCP5/6-specific extensions, while multiple copies of CDK5RAP2 engage the γ-TuRC in two distinct binding patterns to promote γ-TuRC closure and activation. In the centriole lumen, the microtubule branching factor Augmin tethers a condensed cluster of γ-TuRCs to the centriole wall with defined directional orientation. Centriole-lumenal γ-TuRC-Augmin is protected from degradation during interphase and released in mitosis to aid chromosome alignment. This study provides a unique view on γ-TuRC structure and molecular organization at centrosomes and identifies an important cellular function of centriole-lumenal γ-TuRCs.
Collapse
Affiliation(s)
- Qi Gao
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Florian W Hofer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Sebastian Filbeck
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Bram J A Vermeulen
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Martin Würtz
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Annett Neuner
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | | | - Maja Zezlina
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Cornelia Sala
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Hyesu Shin
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | | | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany.
| | - Stefan Pfeffer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany.
| |
Collapse
|
4
|
Selepe MA. Isoflavone Derivatives as Potential Anticancer Agents: Synthesis and Bioactivity Studies. ChemMedChem 2024; 19:e202400420. [PMID: 39091268 PMCID: PMC11617652 DOI: 10.1002/cmdc.202400420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/04/2024]
Abstract
Isoflavones are phenolic natural compounds with a C6C3C6 framework. They possess a plethora of biological activities that are associated with putative benefits to human health. In particular, the cancer chemopreventive and chemotherapeutic potential of isoflavones has attracted the interest of researchers. Several isoflavone derivatives have been synthesised and probed for their anticancer activities. The isoflavone analogues are mainly synthesised by molecular hybridisation and other strategies that enable diversification through early or late-stage functionalisation of A-, B- and C-rings of the isoflavones. This has resulted in the discovery of isoflavone analogues with improved antiproliferative activities against several cancer cells and different mechanisms of action. In this review, the synthesis of isoflavone derivatives and their anticancer activity studies are discussed.
Collapse
Affiliation(s)
- Mamoalosi A. Selepe
- Department of Chemistry, Faculty of Natural and Agricultural SciencesUniversity of PretoriaPrivate bag X 20Hatfield0028South Africa
| |
Collapse
|
5
|
Vermeulen BJ, Böhler A, Gao Q, Neuner A, Župa E, Chu Z, Würtz M, Jäkle U, Gruss OJ, Pfeffer S, Schiebel E. γ-TuRC asymmetry induces local protofilament mismatch at the RanGTP-stimulated microtubule minus end. EMBO J 2024; 43:2062-2085. [PMID: 38600243 PMCID: PMC11099078 DOI: 10.1038/s44318-024-00087-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
The γ-tubulin ring complex (γ-TuRC) is a structural template for de novo microtubule assembly from α/β-tubulin units. The isolated vertebrate γ-TuRC assumes an asymmetric, open structure deviating from microtubule geometry, suggesting that γ-TuRC closure may underlie regulation of microtubule nucleation. Here, we isolate native γ-TuRC-capped microtubules from Xenopus laevis egg extract nucleated through the RanGTP-induced pathway for spindle assembly and determine their cryo-EM structure. Intriguingly, the microtubule minus end-bound γ-TuRC is only partially closed and consequently, the emanating microtubule is locally misaligned with the γ-TuRC and asymmetric. In the partially closed conformation of the γ-TuRC, the actin-containing lumenal bridge is locally destabilised, suggesting lumenal bridge modulation in microtubule nucleation. The microtubule-binding protein CAMSAP2 specifically binds the minus end of γ-TuRC-capped microtubules, indicating that the asymmetric minus end structure may underlie recruitment of microtubule-modulating factors for γ-TuRC release. Collectively, we reveal a surprisingly asymmetric microtubule minus end protofilament organisation diverging from the regular microtubule structure, with direct implications for the kinetics and regulation of nucleation and subsequent modulation of microtubules during spindle assembly.
Collapse
Affiliation(s)
- Bram Ja Vermeulen
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Anna Böhler
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Qi Gao
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Annett Neuner
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Erik Župa
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Zhenzhen Chu
- Institut für Genetik, Universität Bonn, Bonn, Germany
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Lymphoma Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Martin Würtz
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Ursula Jäkle
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany
| | | | - Stefan Pfeffer
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany.
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg, Germany.
| |
Collapse
|
6
|
Vottero P, Wang Q, Michalak M, Aminpour M, Tuszynski JA. Computational Analysis and Experimental Testing of the Molecular Mode of Action of Gatastatin and Its Derivatives. Cancers (Basel) 2023; 15:cancers15061714. [PMID: 36980600 PMCID: PMC10046562 DOI: 10.3390/cancers15061714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Given its critical role in cell mitosis, the tubulin γ chain represents a viable chemotherapeutic target to solve the specificity issues associated with targeting α and β tubulin. Since γ tubulin is overexpressed in glioblastoma multiforme (GBM) and some breast lesions, the glaziovianin A derivative gatastatin, presented as a γ-tubulin-specific inhibitor, could yield a successful therapeutic strategy. The present work aims to identify the binding sites and modes of gatastatin and its derivatives through molecular-docking simulations. Computational binding free energy predictions were compared to experimental microscale thermophoresis assay results. The computational simulations did not reveal a strong preference toward γ tubulin, suggesting that further derivatization may be needed to increase its specificity.
Collapse
Affiliation(s)
- Paola Vottero
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 2V2, Canada
| | - Qian Wang
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Maral Aminpour
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 2V2, Canada
| | - Jack Adam Tuszynski
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E9, Canada
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, I-10129 Turin, Italy
- Correspondence:
| |
Collapse
|
7
|
Yuki R, Ikeda Y, Yasutake R, Saito Y, Nakayama Y. SH2D4A promotes centrosome maturation to support spindle microtubule formation and mitotic progression. Sci Rep 2023; 13:2067. [PMID: 36739326 PMCID: PMC9899277 DOI: 10.1038/s41598-023-29362-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/02/2023] [Indexed: 02/06/2023] Open
Abstract
Mitotic progression requires the precise formation of spindle microtubules based on mature centrosomes. During the G2/M transition, centrosome maturation progresses, and associated microtubules bundle to form mitotic spindle fibers and capture the chromosomes for alignment at the cell equator. Mitotic kinases-induced phosphorylation signaling is necessary for these processes. Here, we identified SH2 domain-containing protein 4A (SH2D4A/PPP1R38) as a new mitotic regulator. SH2D4A knockdown delays mitotic progression. The time-lapse imaging analysis showed that SH2D4A specifically contributes to the alignment of chromosomes. The cold treatment assay and microtubule regrowth assay indicated that SH2D4A promotes microtubule nucleation to support kinetochore-microtubule attachment. This may be due to the centrosome maturation by SH2D4A via centrosomal recruitment of pericentriolar material (PCM) such as cep192, γ-tubulin, and PLK1. SH2D4A was found to be a negative regulator of PP1 phosphatase. Consistently, treatment with a PP1 inhibitor rescues SH2D4A-knockdown-induced phenotypes, including the microtubule nucleation and centrosomal recruitment of active PLK1. These results suggest that SH2D4A is involved in PCM recruitment to centrosomes and centrosome maturation through attenuation of PP1 phosphatases, accelerating the spindle formation and supporting mitotic progression.
Collapse
Affiliation(s)
- Ryuzaburo Yuki
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, 5 Misasagi-Nakauchi-cho, Yamashina-ku, Kyoto, 607-8414, Japan.
| | - Yuki Ikeda
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, 5 Misasagi-Nakauchi-cho, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Ryuji Yasutake
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, 5 Misasagi-Nakauchi-cho, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Youhei Saito
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, 5 Misasagi-Nakauchi-cho, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Yuji Nakayama
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, 5 Misasagi-Nakauchi-cho, Yamashina-ku, Kyoto, 607-8414, Japan.
| |
Collapse
|
8
|
An update on the recent advances and discovery of novel tubulin colchicine binding inhibitors. Future Med Chem 2023; 15:73-95. [PMID: 36756851 DOI: 10.4155/fmc-2022-0212] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Microtubules, formed by α- and β-tubulin heterodimer, are considered as a major target to prevent the proliferation of tumor cells. Microtubule-targeted agents have become increasingly effective anticancer drugs. However, due to the relatively sophisticated chemical structure of taxane and vinblastine, their application has faced numerous obstacles. Conversely, the structure of colchicine binding site inhibitors (CBSIs) is much easier to be modified. Moreover, CBSIs have strong antiproliferative effect on multidrug-resistant tumor cells and have become the mainstream research orientation of microtubule-targeted agents. This review focuses mainly on the recent advances of CBSIs during 2017-2022, attempts to depict their biological activities to analyze the structure-activity relationships and offers new perspectives for designing next generation of novel CBSIs.
Collapse
|
9
|
Lafanechère L. The microtubule cytoskeleton: An old validated target for novel therapeutic drugs. Front Pharmacol 2022; 13:969183. [PMID: 36188585 PMCID: PMC9521402 DOI: 10.3389/fphar.2022.969183] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Compounds targeting microtubules are widely used in cancer therapy with a proven efficacy. However, because they also target non-cancerous cells, their administration leads to numerous adverse effects. With the advancement of knowledge on the structure of tubulin, the regulation of microtubule dynamics and their deregulation in pathological processes, new therapeutic strategies are emerging, both for the treatment of cancer and for other diseases, such as neuronal or even heart diseases and parasite infections. In addition, a better understanding of the mechanism of action of well-known drugs such as colchicine or certain kinase inhibitors contributes to the development of these new therapeutic approaches. Nowadays, chemists and biologists are working jointly to select drugs which target the microtubule cytoskeleton and have improved properties. On the basis of a few examples this review attempts to depict the panorama of these recent advances.
Collapse
|
10
|
Hayakawa I, Usui T. Development of Gatastatin G2, a γ-Tubulin-specific Inhibitor. J SYN ORG CHEM JPN 2022. [DOI: 10.5059/yukigoseikyokaishi.80.563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ichiro Hayakawa
- Graduate School of Integrated Basic Sciences, Nihon University
| | - Takeo Usui
- Faculty of Life and Environmental Sciences, University of Tsukuba
| |
Collapse
|
11
|
Tsuchiya K, Goshima G. Microtubule-associated proteins promote microtubule generation in the absence of γ-tubulin in human colon cancer cells. J Cell Biol 2021; 220:e202104114. [PMID: 34779859 PMCID: PMC8598081 DOI: 10.1083/jcb.202104114] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/13/2021] [Accepted: 09/20/2021] [Indexed: 11/29/2022] Open
Abstract
The γ-tubulin complex acts as the predominant microtubule (MT) nucleator that initiates MT formation and is therefore an essential factor for cell proliferation. Nonetheless, cellular MTs are formed after experimental depletion of the γ-tubulin complex, suggesting that cells possess other factors that drive MT nucleation. Here, by combining gene knockout, auxin-inducible degron, RNA interference, MT depolymerization/regrowth assay, and live microscopy, we identified four microtubule-associated proteins (MAPs), ch-TOG, CLASP1, CAMSAPs, and TPX2, which are involved in γ-tubulin-independent MT generation in human colon cancer cells. In the mitotic MT regrowth assay, nucleated MTs organized noncentriolar MT organizing centers (ncMTOCs) in the absence of γ-tubulin. Depletion of CLASP1 or TPX2 substantially delayed ncMTOC formation, suggesting that these proteins might promote MT nucleation in the absence of γ-tubulin. In contrast, depletion of ch-TOG or CAMSAPs did not affect the timing of ncMTOC appearance. CLASP1 also accelerates γ-tubulin-independent MT regrowth during interphase. Thus, MT generation can be promoted by MAPs without the γ-tubulin template.
Collapse
Affiliation(s)
- Kenta Tsuchiya
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Gohta Goshima
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Sugashima Marine Biological Laboratory, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
12
|
Dráber P, Dráberová E. Dysregulation of Microtubule Nucleating Proteins in Cancer Cells. Cancers (Basel) 2021; 13:cancers13225638. [PMID: 34830792 PMCID: PMC8616210 DOI: 10.3390/cancers13225638] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The dysfunction of microtubule nucleation in cancer cells changes the overall cytoskeleton organization and cellular physiology. This review focuses on the dysregulation of the γ-tubulin ring complex (γ-TuRC) proteins that are essential for microtubule nucleation. Recent research on the high-resolution structure of γ-TuRC has brought new insight into the microtubule nucleation mechanism. We discuss the effect of γ-TuRC protein overexpression on cancer cell behavior and new drugs directed to γ-tubulin that may offer a viable alternative to microtubule-targeting agents currently used in cancer chemotherapy. Abstract In cells, microtubules typically nucleate from microtubule organizing centers, such as centrosomes. γ-Tubulin, which forms multiprotein complexes, is essential for nucleation. The γ-tubulin ring complex (γ-TuRC) is an efficient microtubule nucleator that requires additional centrosomal proteins for its activation and targeting. Evidence suggests that there is a dysfunction of centrosomal microtubule nucleation in cancer cells. Despite decades of molecular analysis of γ-TuRC and its interacting factors, the mechanisms of microtubule nucleation in normal and cancer cells remains obscure. Here, we review recent work on the high-resolution structure of γ-TuRC, which brings new insight into the mechanism of microtubule nucleation. We discuss the effects of γ-TuRC protein dysregulation on cancer cell behavior and new compounds targeting γ-tubulin. Drugs inhibiting γ-TuRC functions could represent an alternative to microtubule targeting agents in cancer chemotherapy.
Collapse
|
13
|
Rayevsky A, Sharifi M, Samofalova D, Demchuk O, Karpov P, Blume Y. In silico mechanistic model of microtubule assembly inhibition by selective chromone derivatives. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
14
|
Trapika IGMGSC, Liu XT, Chung LH, Lai F, Xie C, Zhao Y, Cui S, Chen J, Tran C, Wang Q, Zhang S, Don AS, Li GQ, Hanrahan JR, Qi Y. Ceramide Regulates Anti-Tumor Mechanisms of Erianin in Androgen-Sensitive and Castration-Resistant Prostate Cancers. Front Oncol 2021; 11:738078. [PMID: 34604081 PMCID: PMC8484793 DOI: 10.3389/fonc.2021.738078] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/02/2021] [Indexed: 01/09/2023] Open
Abstract
Prostate cancer is the second most prevalent malignancy worldwide. In the early stages, the development of prostate cancer is dependent on androgens. Over time with androgen deprivation therapy, 20% of prostate cancers progress to a castration-resistant form. Novel treatments for prostate cancers are still urgently needed. Erianin is a plant-derived bibenzyl compound. We report herein that erianin exhibits anti-tumor effects in androgen-sensitive and castration-resistant prostate cancer cells through different mechanisms. Erianin induces endoplasmic reticulum stress-associated apoptosis in androgen-sensitive prostate cancer cells. It also triggers pro-survival autophagic responses, as inhibition of autophagy predisposes to apoptosis. In contrast, erianin fails to induce apoptosis in castration-resistant prostate cancer cells. Instead, it results in cell cycle arrest at the M phase. Mechanistically, C16 ceramide dictates differential responses of androgen-sensitive and castration-resistant prostate cancer cells to erianin. Erianin elevates C16 ceramide level in androgen-sensitive but not castration-resistant prostate cancer cells. Overexpression of ceramide synthase 5 that specifically produces C16 ceramide enables erianin to induce apoptosis in castration-resistant prostate cancer cells. Our study provides both experimental evidence and mechanistic data showing that erianin is a potential treatment option for prostate cancers.
Collapse
Affiliation(s)
- I Gusti Md Gde Surya C. Trapika
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia,School of Pharmacy, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Xin Tracy Liu
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia
| | - Long Hoa Chung
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia
| | - Felcia Lai
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia,School of Pharmacy, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Chanlu Xie
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia,Chinese Medicine Anti-Cancer Evaluation Program, Central Clinical School, University of Sydney, Camperdown, NSW, Australia
| | - Yang Zhao
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaohui Cui
- Key Laboratory of Biotechnology and Biorescources Utilization of Ministry of Education, Dalian Minzu University, Dalian, China
| | - Jinbiao Chen
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia
| | - Collin Tran
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia
| | - Qian Wang
- Translational Cancer Metabolism Laboratory, School of Medical Sciences and Prince of Wales Clinical School, UNSW, Sydney, NSW, Australia
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Biorescources Utilization of Ministry of Education, Dalian Minzu University, Dalian, China
| | - Anthony S. Don
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia,School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia
| | - George Qian Li
- School of Pharmacy, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Jane R. Hanrahan
- School of Pharmacy, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia,*Correspondence: Yanfei Qi, ; Jane R. Hanrahan,
| | - Yanfei Qi
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia,*Correspondence: Yanfei Qi, ; Jane R. Hanrahan,
| |
Collapse
|
15
|
Ebisu H, Shintani K, Chinen T, Nagumo Y, Shioda S, Hatanaka T, Sakakura A, Hayakawa I, Kigoshi H, Usui T. Dual Inhibition of γ-Tubulin and Plk1 Induces Mitotic Cell Death. Front Pharmacol 2021; 11:620185. [PMID: 33584305 PMCID: PMC7878676 DOI: 10.3389/fphar.2020.620185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/30/2020] [Indexed: 11/25/2022] Open
Abstract
α/β-Tubulin inhibitors that alter microtubule (MT) dynamics are commonly used in cancer therapy, however, these inhibitors also cause severe side effects such as peripheral neuropathy. γ-Tubulin is a possible target as antitumor drugs with low side effects, but the antitumor effect of γ-tubulin inhibitors has not been reported yet. In this study, we verified the antitumor activity of gatastatin, a γ-tubulin specific inhibitor. The cytotoxicity of gatastatin was relatively weak compared with that of the conventional MT inhibitors, paclitaxel and vinblastine. To improve the cytotoxicity, we screened the chemicals that improve the effects of gatastatin and found that BI 2536, a Plk1 inhibitor, greatly increases the cytotoxicity of gatastatin. Co-treatment with gatastatin and BI 2536 arrested cell cycle progression at mitosis with abnormal spindles. Moreover, mitotic cell death induced by the combined treatment was suppressed by the Mps1 inhibitor, reversine. These findings suggest that co-treatment with Plk1 and γ-tubulin inhibitors causes spindle assembly checkpoint-dependent mitotic cell death by impairing centrosome functions. These results raise the possibility of Plk1 and γ-tubulin inhibitor co-treatment as a novel cancer chemotherapy.
Collapse
Affiliation(s)
- Haruna Ebisu
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Kana Shintani
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Takumi Chinen
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,Department of Molecular Genetics, Division of Centrosome Biology, National Institute of Genetics, Mishima, Japan.,Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Yoko Nagumo
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Shuya Shioda
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| | - Taisei Hatanaka
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Akira Sakakura
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Ichiro Hayakawa
- Graduate School of Integrated Basic Sciences, Nihon University, Tokyo, Japan
| | - Hideo Kigoshi
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| | - Takeo Usui
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
16
|
Xia LY, Zhang YL, Yang R, Wang ZC, Lu YD, Wang BZ, Zhu HL. Tubulin Inhibitors Binding to Colchicine-Site: A Review from 2015 to 2019. Curr Med Chem 2021; 27:6787-6814. [PMID: 31580244 DOI: 10.2174/0929867326666191003154051] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/25/2019] [Accepted: 08/22/2019] [Indexed: 11/22/2022]
Abstract
Due to the three domains of the colchicine-site which is conducive to the combination with small molecule compounds, colchicine-site on the tubulin has become a common target for antitumor drug development, and accordingly, a large number of tubulin inhibitors binding to the colchicine-site have been reported and evaluated over the past years. In this study, tubulin inhibitors targeting the colchicine-site and their application as antitumor agents were reviewed based on the literature from 2015 to 2019. Tubulin inhibitors were classified into ten categories according to the structural features, including colchicine derivatives, CA-4 analogs, chalcone analogs, coumarin analogs, indole hybrids, quinoline and quinazoline analogs, lignan and podophyllotoxin derivatives, phenothiazine analogs, N-heterocycle hybrids and others. Most of them displayed potent antitumor activity, including antiproliferative effects against Multi-Drug-Resistant (MDR) cell lines and antivascular properties, both in vitro and in vivo. In this review, the design, synthesis and the analysis of the structure-activity relationship of tubulin inhibitors targeting the colchicine-site were described in detail. In addition, multi-target inhibitors, anti-MDR compounds, and inhibitors bearing antitumor activity in vivo are further listed in tables to present a clear picture of potent tubulin inhibitors, which could be beneficial for medicinal chemistry researchers.
Collapse
Affiliation(s)
- Lin-Ying Xia
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Ya-Liang Zhang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Rong Yang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Zhong-Chang Wang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Ya-Dong Lu
- Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Bao-Zhong Wang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Hai-Liang Zhu
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China
| |
Collapse
|
17
|
Zhu T, Wang SH, Li D, Wang SY, Liu X, Song J, Wang YT, Zhang SY. Progress of tubulin polymerization activity detection methods. Bioorg Med Chem Lett 2021; 37:127698. [PMID: 33468346 DOI: 10.1016/j.bmcl.2020.127698] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/05/2020] [Accepted: 11/14/2020] [Indexed: 12/13/2022]
Abstract
Tubulin, an important target in tumor therapy, is one of the hotspots in the field of antineoplastic drugs in recent years, and it is of great significance to design and screen new inhibitors for this target. Natural products and chemical synthetic drugs are the main sources of tubulin inhibitors. However, due to the variety of compound structure types, it has always been difficult for researchers to screen out polymerization inhibitors with simple operation, high efficiency and low cost. A large number of articles have reported the screening methods of tubulin inhibitors and their biological activity. In this article, the biological activity detection methods of tubulin polymerization inhibitors are reviewed. Thus, it provides a theoretical basis for the further study of tubulin polymerization inhibitors and the selection of methods for tubulin inhibitors.
Collapse
Affiliation(s)
- Ting Zhu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Sheng-Hui Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Dong Li
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shu-Yu Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xu Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Ya-Ting Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
18
|
Corvaisier M, Alvarado-Kristensson M. Non-Canonical Functions of the Gamma-Tubulin Meshwork in the Regulation of the Nuclear Architecture. Cancers (Basel) 2020; 12:cancers12113102. [PMID: 33114224 PMCID: PMC7690915 DOI: 10.3390/cancers12113102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/17/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The appearance of a cell is connected to its function. For example, the fusiform of smooth muscle cells is adapted to facilitate muscle contraction, the lobed nucleus in white blood cells assists with the migratory behavior of these immune cells, and the condensed nucleus in sperm aids in their swimming efficiency. Thus, changes in appearance have been used for decades by doctors as a diagnostic method for human cancers. Here, we summarize our knowledge of how a cell maintains the shape of the nuclear compartment. Specifically, we discuss the role of a novel protein meshwork, the gamma-tubulin meshwork, in the regulation of nuclear morphology and as a therapeutic target against cancer. Abstract The nuclear architecture describes the organization of the various compartments in the nucleus of eukaryotic cells, where a plethora of processes such as nucleocytoplasmic transport, gene expression, and assembly of ribosomal subunits occur in a dynamic manner. During the different phases of the cell cycle, in post-mitotic cells and after oncogenic transformation, rearrangements of the nuclear architecture take place, and, among other things, these alterations result in reorganization of the chromatin and changes in gene expression. A member of the tubulin family, γtubulin, was first identified as part of a multiprotein complex that allows nucleation of microtubules. However, more than a decade ago, γtubulin was also characterized as a nuclear protein that modulates several crucial processes that affect the architecture of the nucleus. This review presents the latest knowledge regarding changes that arise in the nuclear architecture of healthy cells and under pathological conditions and, more specifically, considers the particular involvement of γtubulin in the modulation of the biology of the nuclear compartment.
Collapse
|
19
|
Bucko PJ, Garcia I, Manocha R, Bhat A, Wordeman L, Scott JD. Gravin-associated kinase signaling networks coordinate γ-tubulin organization at mitotic spindle poles. J Biol Chem 2020; 295:13784-13797. [PMID: 32732289 PMCID: PMC7535905 DOI: 10.1074/jbc.ra120.014791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/21/2020] [Indexed: 01/16/2023] Open
Abstract
Mitogenic signals that regulate cell division often proceed through multienzyme assemblies within defined intracellular compartments. The anchoring protein Gravin restricts the action of mitotic kinases and cell-cycle effectors to defined mitotic structures. In this report we discover that genetic deletion of Gravin disrupts proper accumulation and asymmetric distribution of γ-tubulin during mitosis. We utilize a new precision pharmacology tool, Local Kinase Inhibition, to inhibit the Gravin binding partner polo-like kinase 1 at spindle poles. Using a combination of gene-editing approaches, quantitative imaging, and biochemical assays, we provide evidence that disruption of local polo-like kinase 1 signaling underlies the γ-tubulin distribution defects observed with Gravin loss. Our study uncovers a new role for Gravin in coordinating γ-tubulin recruitment during mitosis and illuminates the mechanism by which signaling enzymes regulate this process at a distinct subcellular location.
Collapse
Affiliation(s)
- Paula J Bucko
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Irvin Garcia
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Ridhima Manocha
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Akansha Bhat
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
20
|
Shintani K, Ebisu H, Mukaiyama M, Hatanaka T, Chinen T, Takao D, Nagumo Y, Sakakura A, Hayakawa I, Usui T. Structure Optimization of Gatastatin for the Development of γ-Tubulin-Specific Inhibitor. ACS Med Chem Lett 2020; 11:1125-1129. [PMID: 32550991 DOI: 10.1021/acsmedchemlett.9b00526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/30/2020] [Indexed: 02/02/2023] Open
Abstract
Gatastatin (O 7-benzyl glaziovianin A) is a γ-tubulin-specific inhibitor that is used to investigate γ-tubulin function in cells. We have previously reported that the unsubstituted phenyl ring of the O 7-benzyl group in gatastatin is important for γ-tubulin inhibition. To obtain further structural information regarding γ-tubulin inhibition, we synthesized several gatastatin derivatives containing a fixed O 7-benzyl moiety. Modifications of the B-ring resulted in drastic decrease in cytotoxicity, abnormal spindle formation activity, and inhibition of microtubule (MT) nucleation. In contrast, various O 6-alkylated gatastatin derivatives showed potent cytotoxicity, induced abnormal spindle formation, and inhibited MT nucleation. We had previously reported that O 6-benzyl glaziovianin A is a potent α/β-tubulin inhibitor; thus, these new results suggest that the O 6-position restricts affinity for α/β- and γ-tubulin. Considering that an O 7-benzyl group increases specificity for γ-tubulin, more potent and specific γ-tubulin inhibitors can be generated through O 6-modifications of gatastatin.
Collapse
Affiliation(s)
| | | | | | - Taisei Hatanaka
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Takumi Chinen
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Daisuke Takao
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | | | - Akira Sakakura
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Ichiro Hayakawa
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | | |
Collapse
|
21
|
Alvarado-Kristensson M. Choreography of the centrosome. Heliyon 2020; 6:e03238. [PMID: 31989056 PMCID: PMC6970175 DOI: 10.1016/j.heliyon.2020.e03238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/31/2022] Open
Abstract
More than a century ago, the centrosome was discovered and described as “the true division organ of the cell”. Electron microscopy revealed that a centrosome is an amorphous structure or pericentriolar protein matrix that surrounds a pair of well-organized centrioles. Today, the importance of the centrosome as a microtubule-organizing center and coordinator of the mitotic spindle is questioned, because centrioles are absent in up to half of all known eukaryotic species, and various mechanisms for acentrosomal microtubule nucleation have been described. This review recapitulates the known functions of centrosome movements in cellular homeostasis and discusses knowledge gaps in this field.
Collapse
Affiliation(s)
- Maria Alvarado-Kristensson
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, SE-20502, Sweden
| |
Collapse
|
22
|
Jiao JB, Wang GZ, Hu XL, Zang Y, Maisonneuve S, Sedgwick AC, Sessler JL, Xie J, Li J, He XP, Tian H. Cyclodextrin-Based Peptide Self-Assemblies (Spds) That Enhance Peptide-Based Fluorescence Imaging and Antimicrobial Efficacy. J Am Chem Soc 2020; 142:1925-1932. [PMID: 31884796 DOI: 10.1021/jacs.9b11207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
As a result of their high specificity for their corresponding biological targets, peptides have shown significant potential in a range of diagnostic and therapeutic applications. However, their widespread use has been limited by their minimal cell permeability and stability in biological milieus. We describe here a hepta-dicyanomethylene-4H-pyran appended β-cyclodextrin (DCM7-β-CD) that acts as a delivery enhancing "host" for 1-bromonaphthalene-modified peptides, as demonstrated with peptide probes P1-P4. Interaction between the fluorescent peptides P1-P3 and DCM7-β-CD results in the hierarchical formation of unique supramolecular architectures, which we term supramolecular-peptide-dots (Spds). Each Spd (Spd-1, Spd-2, and Spd-3) was found to facilitate the intracellular delivery of the constituent fluorescent probes (P1-P3), thus allowing spatiotemporal imaging of an apoptosis biomarker (caspase-3) and mitosis. Spd-4, incorporating the antimicrobial peptide P4, was found to provide an enhanced therapeutic benefit against both Gram-positive and Gram-negative bacteria relative to P4 alone. In addition, a fluorescent Spd-4 was prepared, which revealed greater bacterial cellular uptake compared to the peptide alone (P4-FITC) in E. coli. (ATCC 25922) and S. aureus (ATCC 25923). This latter observation supports the suggestion that the Spd platform reported here has the ability to facilitate the delivery of a therapeutic peptide and provides an easy-to-implement strategy for enhancing the antimicrobial efficacy of known therapeutic peptides. The present findings thus serve to highlight a new and effective supramolecular delivery approach that is potentially generalizable to overcome limitations associated with functional peptides.
Collapse
Affiliation(s)
- Jin-Biao Jiao
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , P. R. China.,Université Paris-Saclay, ENS Paris-Saclay, CNRS, PPSM , 61 av President Wilson , F-94235 Cachan , France
| | - Guan-Zhen Wang
- National Center for Drug Screening, State Key Laboratory of Drug Research Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Rd. , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No. 19A Yuquan Rd. , Beijing 100049 , P. R. China
| | - Xi-Le Hu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , P. R. China
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Rd. , Shanghai 201203 , P. R. China
| | - Stéphane Maisonneuve
- Université Paris-Saclay, ENS Paris-Saclay, CNRS, PPSM , 61 av President Wilson , F-94235 Cachan , France
| | - Adam C Sedgwick
- Department of Chemistry , The University of Texas at Austin , 105 East 24th Street-A5300 , Austin , Texas 78712-1224 , United States
| | - Jonathan L Sessler
- Department of Chemistry , The University of Texas at Austin , 105 East 24th Street-A5300 , Austin , Texas 78712-1224 , United States
| | - Juan Xie
- Université Paris-Saclay, ENS Paris-Saclay, CNRS, PPSM , 61 av President Wilson , F-94235 Cachan , France
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Rd. , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No. 19A Yuquan Rd. , Beijing 100049 , P. R. China.,Open Studio for Druggability Research of Marine Natural Products Pilot National Laboratory for Marine Science and Technology (Qingdao) , 1 Wenhai Rd. , Aoshanwei , Jimo, Qingdao 266237 , P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , P. R. China
| | - He Tian
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , P. R. China
| |
Collapse
|
23
|
Liu P, Zupa E, Neuner A, Böhler A, Loerke J, Flemming D, Ruppert T, Rudack T, Peter C, Spahn C, Gruss OJ, Pfeffer S, Schiebel E. Insights into the assembly and activation of the microtubule nucleator γ-TuRC. Nature 2019; 578:467-471. [DOI: 10.1038/s41586-019-1896-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023]
|
24
|
Niu L, Yang J, Yan W, Yu Y, Zheng Y, Ye H, Chen Q, Chen L. Reversible binding of the anticancer drug KXO1 (tirbanibulin) to the colchicine-binding site of β-tubulin explains KXO1's low clinical toxicity. J Biol Chem 2019; 294:18099-18108. [PMID: 31628188 DOI: 10.1074/jbc.ra119.010732] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/14/2019] [Indexed: 02/05/2023] Open
Abstract
KXO1 (tirbanibulin or KX2-391) is as a non-ATP-competitive inhibitor of SRC proto-oncogene nonreceptor tyrosine kinase (SRC) and is being clinically investigated for the management of various cancers and actinic keratosis. Recently, KXO1 has also been shown to strongly inhibit tubulin. Interestingly, unlike conventional tubulin-targeting drugs, KXO1 has exhibited low toxicity in preclinical and clinical studies, but the reason for this remains elusive, as are the KXO1-binding site and other details of the interaction of KXO1 with tubulin. Here, cell-based experiments revealed that KXO1 induces tubulin depolymerization and G2/M phase cell cycle arrest at low nanomolar concentrations, similar to colchicine, used as a positive control. Results from biochemical experiments, including an N,N-ethylenebis(iodoacetamide) competition assay, disclosed that KXO1 binds to the colchicine-binding site on β-tubulin, further confirmed by the crystal structure of the tubulin-KXO1 complex at 2.5-Å resolution. A high-quality electron density map of the crystallographic data enabled us to unambiguously determine the position and orientation of KXO1 in the colchicine-binding site, revealing the detailed interactions between KXO1 and tubulin. We also found that KXO1 binds reversibly to purified tubulin, induces a totally reversible cellular effect (G2/M cell cycle arrest), and possesses no cellular toxicity 5 days after drug washout, explaining KXO1's low toxicity. In summary, we show that KXO1 binds to the colchicine-binding site of tubulin and resolved the crystal structure of the tubulin-KXO1 complex. Importantly, KXO1's reversible binding to tubulin explains its clinically low toxicity, an insight that could guide further clinical applications of KXO1.
Collapse
Affiliation(s)
- Lu Niu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Yamei Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Yunhua Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China
| | - Qiang Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611730, People's Republic of China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy and Cancer, Chengdu 610041, China.
| |
Collapse
|
25
|
Xia LY, Yang R, Zhang YL, Chu YC, Qi YL, Man RJ, Wang ZC, Wang BZ, Zhu HL. Design, synthesis, and biological evaluation of 2,3-diphenyl-cycloalkyl pyrazole derivatives as potential tubulin polymerization inhibitors. Chem Biol Drug Des 2019; 94:1894-1904. [PMID: 31106514 DOI: 10.1111/cbdd.13565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 01/06/2023]
Abstract
Several novel cycloalkyl-fused 2,3-diaryl pyrazole derivatives were designed, synthesized, and evaluated as potential anti-tubulin agents. Compound A10 exhibited the most potent antiproliferative activity against a panel of cancer lines (IC50 = 0.78-2.42 μM) and low cytotoxicity against 293T & L02 (CC50 values of 131.74 and 174.89 μM, respectively). Moreover, A10 displayed inhibition of tubulin polymerization in vitro, arrested the G2/M phase of the cell cycle, changed morphology of tubulin, increased intracellular reactive oxygen species, and induced apoptosis of HeLa cells. Docking simulation and 3D-QSAR models were performed to elaborate on the anti-tubulin mechanism of the derivatives. The inhibition of monoclonal colony formation provided more intuitional data to verify the possibility of A10 as a novel tubulin assembling inhibitor.
Collapse
Affiliation(s)
- Lin-Ying Xia
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Rong Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ya-Liang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yi-Chun Chu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ya-Lin Qi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ruo-Jun Man
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Guangxi Biological Polysaccharide Separation, Purification and Modification Research Platform, Guangxi University for Nationalities, Nanning, China
| | - Zhong-Chang Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Bao-Zhong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
26
|
Abstract
Microtubules are cytoskeletal filaments essential for numerous aspects of cell physiology. They are polarized polymeric tubes with a fast growing plus end and a slow growing minus end. In this Cell Science at a Glance article and the accompanying poster, we review the current knowledge on the dynamics and organization of microtubule minus ends. Several factors, including the γ-tubulin ring complex, CAMSAP/Patronin, ASPM/Asp, SPIRAL2 (in plants) and the KANSL complex recognize microtubule minus ends and regulate their nucleation, stability and interactions with partners, such as microtubule severing enzymes, microtubule depolymerases and protein scaffolds. Together with minus-end-directed motors, these microtubule minus-end targeting proteins (-TIPs) also control the formation of microtubule-organizing centers, such as centrosomes and spindle poles, and mediate microtubule attachment to cellular membrane structures, including the cell cortex, Golgi complex and the cell nucleus. Structural and functional studies are starting to reveal the molecular mechanisms by which dynamic -TIP networks control microtubule minus ends.
Collapse
Affiliation(s)
- Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland .,University of Basel, Biozentrum, CH-4056 Basel, Switzerland
| |
Collapse
|
27
|
Aberrant methylation status of SPG20 promoter in hepatocellular carcinoma: A potential tumor metastasis biomarker. Cancer Genet 2019; 233-234:48-55. [PMID: 31109594 DOI: 10.1016/j.cancergen.2019.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/01/2019] [Accepted: 04/09/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE The aim of this study is to analyze the methylation levels of SPG20 promotor region and explore the association between the methylation levels and clinical features in hepatocellular carcinoma (HCC). MATERIALS AND METHODS We collected paired of HCC and adjacent non-cancerous tissues (ANT) from 160 HCC patients and analyze the methylation levels through MassARRAY Analyzer 4. The statistical calculations were performed using SPSS version 22.0. Real-time-quantification PCR was performed to assess expression levels of SPG20 in HCC cell lines. Wound healing assay and transwell assay was used to measure cell migration capacity. RESULT We found that mean methylation level of SPG20 in tumor tissues was significantly higher than that in ANT (7.3% vs. 16.2%, P<0.0013). There was a significantly negative correlation between expression level and methylation level of SPG20 (P<0.01). In addition, the methylation levels in HCC were correlated with age and HBV infection. Meanwhile, micro-satellite tumors (P = 0.016) and tumor number (P = 0.018) was found significantly associated with increased methylation levels of several CpG sites and the mean levels of SPG20 promotor in ANT. In addtion, the capacity of cell migration was significantly enhanced in SPG20 knock-down HCC cells. CONCLUSION The hypermethylation status of SPG20 gene promoter is significantly associated with intra-hepatic metastasis and contribute to HCC metastasis.
Collapse
|
28
|
TUBG1 missense variants underlying cortical malformations disrupt neuronal locomotion and microtubule dynamics but not neurogenesis. Nat Commun 2019; 10:2129. [PMID: 31086189 PMCID: PMC6513894 DOI: 10.1038/s41467-019-10081-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 04/05/2019] [Indexed: 01/13/2023] Open
Abstract
De novo heterozygous missense variants in the γ-tubulin gene TUBG1 have been linked to human malformations of cortical development associated with intellectual disability and epilepsy. Here, we investigated through in-utero electroporation and in-vivo studies, how four of these variants affect cortical development. We show that TUBG1 mutants affect neuronal positioning, disrupting the locomotion of new-born neurons but without affecting progenitors’ proliferation. We further demonstrate that pathogenic TUBG1 variants are linked to reduced microtubule dynamics but without major structural nor functional centrosome defects in subject-derived fibroblasts. Additionally, we developed a knock-in Tubg1Y92C/+ mouse model and assessed consequences of the mutation. Although centrosomal positioning in bipolar neurons is correct, they fail to initiate locomotion. Furthermore, Tubg1Y92C/+ animals show neuroanatomical and behavioral defects and increased epileptic cortical activity. We show that Tubg1Y92C/+ mice partially mimic the human phenotype and therefore represent a relevant model for further investigations of the physiopathology of cortical malformations. New mutations and genes associated with malformations of cortical development keep being identified, yet there is little known about the underlying cellular mechanisms controlling these impairments. Here, authors generate and characterize a heterozygous TUBG1 knock-in mouse model bearing one of these known mutations and show that TUBG1 mutation leads to the miss-positioning of neurons in the cortical wall due to migration, because of defective microtubules dynamics, and not proliferation defects during corticogenesis.
Collapse
|
29
|
Kostrhunova H, Zajac J, Novohradsky V, Kasparkova J, Malina J, Aldrich-Wright JR, Petruzzella E, Sirota R, Gibson D, Brabec V. A Subset of New Platinum Antitumor Agents Kills Cells by a Multimodal Mechanism of Action Also Involving Changes in the Organization of the Microtubule Cytoskeleton. J Med Chem 2019; 62:5176-5190. [PMID: 31030506 DOI: 10.1021/acs.jmedchem.9b00489] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The substitution inert platinum agent [Pt(1 S,2 S-diaminocyclohexane)(5,6-dimethyl-1,10-phenanthroline)]2+ (56MeSS, 5) is a potent cytotoxic metallodrug. In contrast to conventional cisplatin or oxaliplatin, the mechanism of action (MoA) of 5 is fundamentally different. However, details of the mechanism by which the 5,6-dimethyl-1,10-phenanthroline ligand contributes to the cytotoxicity of 5 and its derivatives have not been sufficiently clarified so far. Here, we show that 5 and its Pt(IV) derivatives exhibit an intriguing potency in the triple-negative breast cancer cells MDA-MB-231. Moreover, we show that the Pt(IV) derivatives of 5 act by multimodal MoA resulting in the global biological effects, that is, they damage nuclear DNA, reduce the mitochondrial membrane potential, induce the epigenetic processes, and last but not least, the data provide evidence that changes in the organization of cytoskeleton networks are functionally important for 5 and its derivatives, in contrast to clinically used platinum cytostatics, to kill cancer cells.
Collapse
Affiliation(s)
- Hana Kostrhunova
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Juraj Zajac
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Vojtech Novohradsky
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Jana Kasparkova
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Jaroslav Malina
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Janice R Aldrich-Wright
- School of Science and Health , Western Sydney University , Penrith South DC 1797 , NSW , Australia
| | - Emanuele Petruzzella
- Institute for Drug Research, School of Pharmacy , The Hebrew University , Jerusalem 91120 , Israel
| | - Roman Sirota
- Institute for Drug Research, School of Pharmacy , The Hebrew University , Jerusalem 91120 , Israel
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy , The Hebrew University , Jerusalem 91120 , Israel
| | - Viktor Brabec
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| |
Collapse
|
30
|
Traversi G, Staid DS, Fiore M, Percario Z, Trisciuoglio D, Antonioletti R, Morea V, Degrassi F, Cozzi R. A novel resveratrol derivative induces mitotic arrest, centrosome fragmentation and cancer cell death by inhibiting γ-tubulin. Cell Div 2019; 14:3. [PMID: 31007707 PMCID: PMC6457039 DOI: 10.1186/s13008-019-0046-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/23/2019] [Indexed: 12/31/2022] Open
Abstract
Background Resveratrol and its natural stilbene-containing derivatives have been extensively investigated as potential chemotherapeutic agents. The synthetic manipulation of the stilbene scaffold has led to the generation of new analogues with improved anticancer activity and better bioavailability. In the present study we investigated the anticancer activity of a novel trimethoxystilbene derivative (3,4,4'-trimethoxylstilbene), where two methoxyl groups are adjacent on the benzene ring (ortho configuration), and compared its activity to 3,5,4'-trimethoxylstilbene, whose methoxyl groups are in meta configuration. Results We provide evidence that the presence of the two methoxyl groups in ortho configuration renders 3,4,4'-trimethoxystilbene more efficient than the meta isomer in inhibiting cell proliferation and producing apoptotic death in colorectal cancer cells. Confocal microscopy of α- and γ-tubulin staining shows that the novel compound strongly depolymerizes the mitotic spindle and produces fragmentation of the pericentrosomal material. Computer assisted docking studies indicate that both molecules potentially interact with γ-tubulin, and that 3,4,4'-trimethoxystilbene is likely to establish stronger interactions with the protein. Conclusions These findings demonstrate the ortho configuration confers higher specificity for γ-tubulin with respect to α-tubulin on 3,4,4' trimethoxystilbene, allowing it to be defined as a new γ-tubulin inhibitor. A strong interaction with γ-tubulin might be a defining feature of molecules with high anticancer activity, as shown for the 3,4,4' isomer.
Collapse
Affiliation(s)
| | - David Sasah Staid
- 2Department of Biochemical Sciences 'A. Rossi Fanelli', Sapienza University of Rome, Rome, Italy
| | - Mario Fiore
- 3Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), c/o Sapienza University of Rome, Rome, Italy
| | - Zulema Percario
- 1Department of Science, University of "Roma Tre", Rome, Italy
| | - Daniela Trisciuoglio
- 3Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), c/o Sapienza University of Rome, Rome, Italy.,4Preclinical Models and New Therapeutic Agents Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Roberto Antonioletti
- 3Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), c/o Sapienza University of Rome, Rome, Italy
| | - Veronica Morea
- 3Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), c/o Sapienza University of Rome, Rome, Italy
| | - Francesca Degrassi
- 3Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), c/o Sapienza University of Rome, Rome, Italy
| | - Renata Cozzi
- 1Department of Science, University of "Roma Tre", Rome, Italy
| |
Collapse
|
31
|
Hayakawa I, Kigoshi H, Shioda S, Chinen T, Usui T. Structure–Activity Relationship Study of Gatastatin Based on the Topliss Tree Approach. HETEROCYCLES 2019. [DOI: 10.3987/com-18-s(f)16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
32
|
Microtubule nucleation by γ-tubulin complexes and beyond. Essays Biochem 2018; 62:765-780. [PMID: 30315097 PMCID: PMC6281477 DOI: 10.1042/ebc20180028] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/05/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022]
Abstract
In this short review, we give an overview of microtubule nucleation within cells. It is nearly 30 years since the discovery of γ-tubulin, a member of the tubulin superfamily essential for proper microtubule nucleation in all eukaryotes. γ-tubulin associates with other proteins to form multiprotein γ-tubulin ring complexes (γ-TuRCs) that template and catalyse the otherwise kinetically unfavourable assembly of microtubule filaments. These filaments can be dynamic or stable and they perform diverse functions, such as chromosome separation during mitosis and intracellular transport in neurons. The field has come a long way in understanding γ-TuRC biology but several important and unanswered questions remain, and we are still far from understanding the regulation of microtubule nucleation in a multicellular context. Here, we review the current literature on γ-TuRC assembly, recruitment, and activation and discuss the potential importance of γ-TuRC heterogeneity, the role of non-γ-TuRC proteins in microtubule nucleation, and whether γ-TuRCs could serve as good drug targets for cancer therapy.
Collapse
|
33
|
Bärenz F, Kschonsak YT, Meyer A, Jafarpour A, Lorenz H, Hoffmann I. Ccdc61 controls centrosomal localization of Cep170 and is required for spindle assembly and symmetry. Mol Biol Cell 2018; 29:3105-3118. [PMID: 30354798 PMCID: PMC6340214 DOI: 10.1091/mbc.e18-02-0115] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Microtubule nucleation was uncovered as a key principle of spindle assembly. However, the mechanistic details about microtubule nucleation and the organization of spindle formation and symmetry are currently being revealed. Here we describe the function of coiled-coil domain containing 61 (Ccdc61), a so far uncharacterized centrosomal protein, in spindle assembly and symmetry. Our data describe that Ccdc61 is required for spindle assembly and precise chromosome alignments in mitosis. Microtubule tip-tracking experiments in the absence of Ccdc61 reveal a clear loss of the intrinsic symmetry of microtubule tracks within the spindle. Furthermore, we show that Ccdc61 controls the centrosomal localization of centrosomal protein of 170 kDa (Cep170), a protein that was shown previously to localize to centrosomes as well as spindle microtubules and promotes microtubule organization and microtubule assembly. Interestingly, selective disruption of Ccdc61 impairs the binding between Cep170 and TANK binding kinase 1, an interaction that is required for microtubule stability. In summary, we have discovered Ccdc61 as a centrosomal protein with an important function in mitotic microtubule organization.
Collapse
Affiliation(s)
- Felix Bärenz
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Yvonne T Kschonsak
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Annalena Meyer
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Aliakbar Jafarpour
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Holger Lorenz
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Ingrid Hoffmann
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| |
Collapse
|
34
|
Gavilan MP, Gandolfo P, Balestra FR, Arias F, Bornens M, Rios RM. The dual role of the centrosome in organizing the microtubule network in interphase. EMBO Rep 2018; 19:embr.201845942. [PMID: 30224411 DOI: 10.15252/embr.201845942] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 11/09/2022] Open
Abstract
Here, we address the regulation of microtubule nucleation during interphase by genetically ablating one, or two, of three major mammalian γ-TuRC-binding factors namely pericentrin, CDK5Rap2, and AKAP450. Unexpectedly, we find that while all of them participate in microtubule nucleation at the Golgi apparatus, they only modestly contribute at the centrosome where CEP192 has a more predominant function. We also show that inhibiting microtubule nucleation at the Golgi does not affect centrosomal activity, whereas manipulating the number of centrosomes with centrinone modifies microtubule nucleation activity of the Golgi apparatus. In centrosome-free cells, inhibition of Golgi-based microtubule nucleation triggers pericentrin-dependent formation of cytoplasmic-nucleating structures. Further depletion of pericentrin under these conditions leads to the generation of individual microtubules in a γ-tubulin-dependent manner. In all cases, a conspicuous MT network forms. Strikingly, centrosome loss increases microtubule number independently of where they were growing from. Our results lead to an unexpected view of the interphase centrosome that would control microtubule network organization not only by nucleating microtubules, but also by modulating the activity of alternative microtubule-organizing centers.
Collapse
Affiliation(s)
- Maria P Gavilan
- Centro Andaluz de Biología Molecular y Medicina Regenerativa CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| | - Pablo Gandolfo
- Centro Andaluz de Biología Molecular y Medicina Regenerativa CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| | - Fernando R Balestra
- Centro Andaluz de Biología Molecular y Medicina Regenerativa CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| | - Francisco Arias
- Centro Andaluz de Biología Molecular y Medicina Regenerativa CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| | | | - Rosa M Rios
- Centro Andaluz de Biología Molecular y Medicina Regenerativa CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| |
Collapse
|
35
|
Alvarado-Kristensson M. γ-tubulin as a signal-transducing molecule and meshwork with therapeutic potential. Signal Transduct Target Ther 2018; 3:24. [PMID: 30221013 PMCID: PMC6137058 DOI: 10.1038/s41392-018-0021-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/23/2018] [Accepted: 05/06/2018] [Indexed: 01/05/2023] Open
Abstract
Knowledge of γ-tubulin is increasing with regard to the cellular functions of this protein beyond its participation in microtubule nucleation. γ-Tubulin expression is altered in various malignancies, and changes in the TUBG1 gene have been found in patients suffering from brain malformations. This review recapitulates the known functions of γ-tubulin in cellular homeostasis and discusses the possible influence of the protein on disease development and cancer.
Collapse
Affiliation(s)
- Maria Alvarado-Kristensson
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, 20502 Sweden
| |
Collapse
|
36
|
Li QR, Jiao JB, Li LL, He XP, Zang Y, James TD, Chen GR, Guo L, Li J. Graphene oxide-enhanced cytoskeleton imaging and mitosis tracking. Chem Commun (Camb) 2018; 53:3373-3376. [PMID: 28265597 DOI: 10.1039/c7cc01019b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Here we show that graphene oxide greatly enhances the imaging ability of a peptide probe that selectively targets microtubules of the cytoskeleton, thus enabling the dynamic tracking of mitosis in live cells.
Collapse
Affiliation(s)
- Qian-Ru Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, P. R. China.
| | - Jin-Biao Jiao
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| | - Li-Li Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd., Shanghai 201203, P. R. China.
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd., Shanghai 201203, P. R. China.
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK
| | - Guo-Rong Chen
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| | - Lin Guo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, P. R. China.
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd., Shanghai 201203, P. R. China.
| |
Collapse
|
37
|
Yang J, Yan W, Yu Y, Wang Y, Yang T, Xue L, Yuan X, Long C, Liu Z, Chen X, Hu M, Zheng L, Qiu Q, Pei H, Li D, Wang F, Bai P, Wen J, Ye H, Chen L. The compound millepachine and its derivatives inhibit tubulin polymerization by irreversibly binding to the colchicine-binding site in β-tubulin. J Biol Chem 2018; 293:9461-9472. [PMID: 29691282 DOI: 10.1074/jbc.ra117.001658] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/19/2018] [Indexed: 02/05/2023] Open
Abstract
Inhibitors that bind to the paclitaxel- or vinblastine-binding sites of tubulin have been part of the pharmacopoeia of anticancer therapy for decades. However, tubulin inhibitors that bind to the colchicine-binding site are not used in clinical cancer therapy, because of their low therapeutic index. To address multidrug resistance to many conventional tubulin-binding agents, numerous efforts have attempted to clinically develop inhibitors that bind the colchicine-binding site. Previously, we have found that millepachine (MIL), a natural chalcone-type small molecule extracted from the plant Millettia pachycarpa, and its two derivatives (MDs) SKLB028 and SKLB050 have potential antitumor activities both in vitro and in vivo However, their cellular targets and mechanisms are unclear. Here, biochemical and cellular experiments revealed that the MDs directly and irreversibly bind β-tubulin. X-ray crystallography of the tubulin-MD structures disclosed that the MDs bind at the tubulin intradimer interface and to the same site as colchicine and that their binding mode is similar to that of colchicine. Of note, MDs inhibited tubulin polymerization and caused G2/M cell-cycle arrest. Comprehensive analysis further revealed that free MIL exhibits an s-cis conformation, whereas MIL in the colchicine-binding site in tubulin adopts an s-trans conformation. Moreover, introducing an α-methyl to MDs to increase the proportion of s-trans conformations augmented MDs' tubulin inhibition activity. Our study uncovers a new class of chalcone-type tubulin inhibitors that bind the colchicine-binding site in β-tubulin and suggests that the s-trans conformation of these compounds may make them more active anticancer agents.
Collapse
Affiliation(s)
- Jianhong Yang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Wei Yan
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Yamei Yu
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Yuxi Wang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Tao Yang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Linlin Xue
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Xue Yuan
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Caofeng Long
- the Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong 523325, China
| | - Zuowei Liu
- the Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong 523325, China
| | - Xiaoxin Chen
- the Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong 523325, China
| | - Mengshi Hu
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Li Zheng
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Qiang Qiu
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Heying Pei
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Dan Li
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Fang Wang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Peng Bai
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Jiaolin Wen
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Haoyu Ye
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| | - Lijuan Chen
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China and
| |
Collapse
|
38
|
Baudoin NC, Cimini D. A guide to classifying mitotic stages and mitotic defects in fixed cells. Chromosoma 2018; 127:215-227. [PMID: 29411093 DOI: 10.1007/s00412-018-0660-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/07/2018] [Accepted: 01/08/2018] [Indexed: 12/12/2022]
Abstract
Cell division is fundamental to life and its perturbation can disrupt organismal development, alter tissue homeostasis, and cause disease. Analysis of mitotic abnormalities provides insight into how certain perturbations affect the fidelity of cell division and how specific cellular structures, molecules, and enzymatic activities contribute to the accuracy of this process. However, accurate classification of mitotic defects is instrumental for correct interpretation of data and formulation of new hypotheses. In this article, we provide guidelines for identifying specific mitotic stages and for classifying normal and deviant mitotic phenotypes. We hope this will clarify confusion about how certain defects are classified and help investigators avoid misnomers, misclassification, and/or misinterpretation, thus leading to a unified and standardized system to classify mitotic defects.
Collapse
Affiliation(s)
- Nicolaas C Baudoin
- Department of Biological Sciences and Biocomplexity Institute, Virginia Tech, 1015 Life Science Circle, Blacksburg, VA, 24061, USA
| | - Daniela Cimini
- Department of Biological Sciences and Biocomplexity Institute, Virginia Tech, 1015 Life Science Circle, Blacksburg, VA, 24061, USA.
| |
Collapse
|
39
|
Hueschen CL, Kenny SJ, Xu K, Dumont S. NuMA recruits dynein activity to microtubule minus-ends at mitosis. eLife 2017; 6. [PMID: 29185983 PMCID: PMC5706958 DOI: 10.7554/elife.29328] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 10/22/2017] [Indexed: 12/14/2022] Open
Abstract
To build the spindle at mitosis, motors exert spatially regulated forces on microtubules. We know that dynein pulls on mammalian spindle microtubule minus-ends, and this localized activity at ends is predicted to allow dynein to cluster microtubules into poles. How dynein becomes enriched at minus-ends is not known. Here, we use quantitative imaging and laser ablation to show that NuMA targets dynactin to minus-ends, localizing dynein activity there. NuMA is recruited to new minus-ends independently of dynein and more quickly than dynactin; both NuMA and dynactin display specific, steady-state binding at minus-ends. NuMA localization to minus-ends involves a C-terminal region outside NuMA’s canonical microtubule-binding domain and is independent of minus-end binders γ-TuRC, CAMSAP1, and KANSL1/3. Both NuMA’s minus-end-binding and dynein-dynactin-binding modules are required to rescue focused, bipolar spindle organization. Thus, NuMA may serve as a mitosis-specific minus-end cargo adaptor, targeting dynein activity to minus-ends to cluster spindle microtubules into poles. Every time a cell divides, it needs to duplicate its DNA and evenly distribute it between the two new ‘daughter’ cells. To move and distribute DNA, the cell builds a large machine called a spindle, which is made of stiff cables called microtubules. Many proteins, including a motor called dynein, help to organize the spindle’s microtubules. One of dynein’s jobs is to cluster all microtubules at the two tips of the spindle, pulling them into shape. Without this clustering, spindles have the wrong shape and structure and can make mistakes when moving DNA. Microtubules have a ‘plus’ end and a ‘minus’ end, and motor proteins usually only travel in one specified direction. Dynein, for example, moves towards the minus end of microtubules, which is where most of the clustering happens. It can form a complex with other proteins that help clustering, one of which is called NuMA. Until now, it was thought that dynein transports NuMA to the minus ends. To test this model, Hueschen et al. studied dividing human cells under a microscope and isolated minus ends with the help of a laser. The experiments showed that, in fact, NuMA gets to minus ends independently of dynein. Once it is on the minus ends, NuMA grabs hold of another protein complex called dynactin, which then gathers dynein. Dynein then pulls the spindles into shape from the minus ends. When NuMA was experimentally removed from the cells, dynein-dynactin complexes were scattered along the entire length of the microtubule instead of pulling specifically on minus-ends, which resulted in disorganized spindles. Thus, where dynein complexes pull determines what spindle shape they build. Hueschen et al. also showed that dynein complexes only pull on minus-ends while the cell divides, which makes sense, because NuMA remains hidden in the cell nucleus for the rest of the time. Together, these results suggest that NuMA makes sure dynein pulls specifically on the minus-ends of the microtubules to tighten the spindle at the right time. A next step will be to test how the mechanical properties of the spindle are changed without dynein pulling on minus-ends. A better knowledge of how different proteins work together to build the spindle and help cells divide can help us understand what goes wrong when cells divide abnormally, as in the case of cancer cells.
Collapse
Affiliation(s)
- Christina L Hueschen
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States.,Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Samuel J Kenny
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Sophie Dumont
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States.,Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, United States.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
40
|
EB1-binding-myomegalin protein complex promotes centrosomal microtubules functions. Proc Natl Acad Sci U S A 2017; 114:E10687-E10696. [PMID: 29162697 DOI: 10.1073/pnas.1705682114] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Control of microtubule dynamics underlies several fundamental processes such as cell polarity, cell division, and cell motility. To gain insights into the mechanisms that control microtubule dynamics during cell motility, we investigated the interactome of the microtubule plus-end-binding protein end-binding 1 (EB1). Via molecular mapping and cross-linking mass spectrometry we identified and characterized a large complex associating a specific isoform of myomegalin termed "SMYLE" (for short myomegalin-like EB1 binding protein), the PKA scaffolding protein AKAP9, and the pericentrosomal protein CDK5RAP2. SMYLE was associated through an evolutionarily conserved N-terminal domain with AKAP9, which in turn was anchored at the centrosome via CDK5RAP2. SMYLE connected the pericentrosomal complex to the microtubule-nucleating complex (γ-TuRC) via Galectin-3-binding protein. SMYLE associated with nascent centrosomal microtubules to promote microtubule assembly and acetylation. Disruption of SMYLE interaction with EB1 or AKAP9 prevented microtubule nucleation and their stabilization at the leading edge of migrating cells. In addition, SMYLE depletion led to defective astral microtubules and abnormal orientation of the mitotic spindle and triggered G1 cell-cycle arrest, which might be due to defective centrosome integrity. As a consequence, SMYLE loss of function had a profound impact on tumor cell motility and proliferation, suggesting that SMYLE might be an important player in tumor progression.
Collapse
|
41
|
Characterization of gamma-tubulin filaments in mammalian cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:158-171. [PMID: 29050966 DOI: 10.1016/j.bbamcr.2017.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/20/2017] [Accepted: 10/13/2017] [Indexed: 11/20/2022]
Abstract
Overexpression of γ-tubulin leads to the formation of filaments, but nothing is known about such filaments with regard to possible presence in cells, structure and probable dynamics. Here, we used mammalian cell lines to investigate the ability of γ-tubulin to form filaments. We found that γ-tubulin produces fibers called γ-tubules in a GTP-dependent manner and that γ-tubules are made up of pericentrin and the γ-tubulin complex proteins 2, 3, 5 and 6. Furthermore, we noted that the number of cells with cytosolic γ-tubules is increased in non-dividing cells. Our experiments showed that γ-tubules are polar structures that have a low regrowth rate compared to microtubules. Also, we observed that γ-tubules were disassembled by treatment with cold, colcemid, citral dimethyl acetal, dimethyl fumarate or mutation of γ-tubulin GTPase domain, but were increased in number by treatment with taxol or by stable expression of the γ-tubulin1-333 GTPase domain. Our results demonstrate that γ-tubulin forms filaments, and such assembly is facilitated by the GTPase domain of γ-tubulin.
Collapse
|
42
|
Semenova I, Gupta D, Usui T, Hayakawa I, Cowan A, Rodionov V. Stimulation of microtubule-based transport by nucleation of microtubules on pigment granules. Mol Biol Cell 2017; 28:1418-1425. [PMID: 28381426 PMCID: PMC5449142 DOI: 10.1091/mbc.e16-08-0571] [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: 08/04/2016] [Revised: 03/27/2017] [Accepted: 03/29/2017] [Indexed: 11/11/2022] Open
Abstract
In Xenopus melanophores, nucleation of microtubules on pigment granules provides a positive feedback loop that enhances their transport to the cell center during pigment aggregation. Microtubule (MT)-based transport can be regulated through changes in organization of MT transport tracks, but the mechanisms that regulate these changes are poorly understood. In Xenopus melanophores, aggregation of pigment granules in the cell center involves their capture by the tips of MTs growing toward the cell periphery, and granule aggregation signals facilitate capture by increasing the number of growing MT tips. This increase could be explained by stimulation of MT nucleation either on the centrosome or on the aggregate of pigment granules that gradually forms in the cell center. We blocked movement of pigment granules to the cell center and compared the MT-nucleation activity of the centrosome in the same cells in two signaling states. We found that granule aggregation signals did not stimulate MT nucleation on the centrosome but did increase MT nucleation activity of pigment granules. Elevation of MT-nucleation activity correlated with the recruitment to pigment granules of a major component of MT-nucleation templates, γ-tubulin, and was suppressed by γ-tubulin inhibitors. We conclude that generation of new MT transport tracks by concentration of the leading pigment granules provides a positive feedback loop that enhances delivery of trailing granules to the cell center.
Collapse
Affiliation(s)
- Irina Semenova
- R. D. Berlin Center for Cell Analysis and Modeling and Department of Cell Biology, UConn Health, Farmington, CT 06030
| | - Dipika Gupta
- R. D. Berlin Center for Cell Analysis and Modeling and Department of Cell Biology, UConn Health, Farmington, CT 06030
| | - Takeo Usui
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan
| | - Ichiro Hayakawa
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Ann Cowan
- R. D. Berlin Center for Cell Analysis and Modeling and Department of Cell Biology, UConn Health, Farmington, CT 06030
| | - Vladimir Rodionov
- R. D. Berlin Center for Cell Analysis and Modeling and Department of Cell Biology, UConn Health, Farmington, CT 06030
| |
Collapse
|
43
|
Hayakawa I, Shioda S, Chinen T, Hatanaka T, Ebisu H, Sakakura A, Usui T, Kigoshi H. Discovery of O 6-benzyl glaziovianin A, a potent cytotoxic substance and a potent inhibitor of α,β-tubulin polymerization. Bioorg Med Chem 2016; 24:5639-5645. [PMID: 27665177 DOI: 10.1016/j.bmc.2016.09.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/09/2016] [Accepted: 09/10/2016] [Indexed: 10/21/2022]
Abstract
We have discovered O6-benzyl glaziovianin A, which showed stronger inhibition of microtubule polymerization (IC50=2.1μM) than known α,β-tubulin inhibitors, such as colchicine and glaziovianin A. Also, we performed competition binding experiments of O6-benzyl glaziovianin A and revealed that O6-benzyl glaziovianin A binds to the colchicine binding site with high affinity. It is interesting that glaziovianin A derivatives change their mode of action in benzylation at the O6 (α,β-tubulin inhibitor) or O7 (γ-tubulin-specific inhibitor) position.
Collapse
Affiliation(s)
- Ichiro Hayakawa
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Shuya Shioda
- Department of Chemistry, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8571, Japan
| | - Takumi Chinen
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| | - Taisei Hatanaka
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Haruna Ebisu
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| | - Akira Sakakura
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Takeo Usui
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan.
| | - Hideo Kigoshi
- Department of Chemistry, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8571, Japan.
| |
Collapse
|
44
|
Farache D, Jauneau A, Chemin C, Chartrain M, Rémy MH, Merdes A, Haren L. Functional Analysis of γ-Tubulin Complex Proteins Indicates Specific Lateral Association via Their N-terminal Domains. J Biol Chem 2016; 291:23112-23125. [PMID: 27660388 DOI: 10.1074/jbc.m116.744862] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Indexed: 11/06/2022] Open
Abstract
Microtubules are nucleated from multiprotein complexes containing γ-tubulin and associated γ-tubulin complex proteins (GCPs). Small complexes (γTuSCs) comprise two molecules of γ-tubulin bound to the C-terminal domains of GCP2 and GCP3. γTuSCs associate laterally into helical structures, providing a structural template for microtubule nucleation. In most eukaryotes γTuSCs associate with additional GCPs (4, 5, and 6) to form the core of the so-called γ-tubulin ring complex (γTuRC). GCPs 2-6 constitute a family of homologous proteins. Previous structural analysis and modeling of GCPs suggest that all family members can potentially integrate into the helical structure. Here we provide experimental evidence for this model. Using chimeric proteins in which the N- and C-terminal domains of different GCPs are swapped, we show that the N-terminal domains define the functional identity of GCPs, whereas the C-terminal domains are exchangeable. FLIM-FRET experiments indicate that GCP4 and GCP5 associate laterally within the complex, and their interaction is mediated by their N-terminal domains as previously shown for γTuSCs. Our results suggest that all GCPs are incorporated into the helix via lateral interactions between their N-terminal domains, whereas the C-terminal domains mediate longitudinal interactions with γ-tubulin. Moreover, we show that binding to γ-tubulin is not essential for integrating into the helical complex.
Collapse
Affiliation(s)
- Dorian Farache
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Alain Jauneau
- Plateforme Imagerie-Microscopie, FR 3450 Pôle de Biotechnologie Végétale, 31326 Castanet-Tolosan, France
| | - Cécile Chemin
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Marine Chartrain
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Marie-Hélène Rémy
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Andreas Merdes
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Laurence Haren
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| |
Collapse
|
45
|
Semenov VV, Tsyganov DV, Semenova MN, Chuprov-Netochin RN, Raihstat MM, Konyushkin LD, Volynchuk PB, Marusich EI, Nazarenko VV, Leonov SV, Kiselyov AS. Efficient Synthesis of Glaziovianin A Isoflavone Series from Dill and Parsley Extracts and Their in Vitro/in Vivo Antimitotic Activity. JOURNAL OF NATURAL PRODUCTS 2016; 79:1429-1438. [PMID: 27100701 DOI: 10.1021/acs.jnatprod.6b00173] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
A concise six-step protocol for the synthesis of isoflavone glaziovianin A (GVA) and its alkoxyphenyl derivatives 9 starting with readily available plant metabolites from dill and parsley seeds was developed. The reaction sequence involved an efficient conversion of the key intermediate epoxides 7 into the respective β-ketoaldehydes 8 followed by their Cu(I)-mediated cyclization into the target series 9. The biological activity of GVA and its derivatives was evaluated using a panel of seven human cancer cell lines and an in vivo sea urchin embryo assay. Both screening platforms confirmed the antimitotic effect of the parent GVA (9cg) and its alkoxy derivatives. Structure-activity relationship studies suggested that compounds 9cd and 9cf substituted with trimethoxy- and dillapiol-derived B-rings, respectively, were less active than the parent 9cg. Of the evaluated human cancer cell lines, the A375 melanoma cell line was the most sensitive to the tested molecules. Notably, the target compounds were not cytotoxic against human peripheral blood mononuclear cells up to 10 μM concentration. Phenotypic readouts from the sea urchin assay unequivocally suggest a direct microtubule-destabilizing effect of isoflavones 9cg, 9cd, and 9cf.
Collapse
Affiliation(s)
- Victor V Semenov
- N. D. Zelinsky Institute of Organic Chemistry, RAS , Leninsky Prospect, 47, 119991, Moscow, Russian Federation
| | - Dmitry V Tsyganov
- N. D. Zelinsky Institute of Organic Chemistry, RAS , Leninsky Prospect, 47, 119991, Moscow, Russian Federation
| | - Marina N Semenova
- Institute of Developmental Biology, RAS , Vavilov Street, 26, 119334, Moscow, Russian Federation
- Chemical Block Ltd. , 3 Kyriacou Matsi, 3723, Limassol, Cyprus
| | - Roman N Chuprov-Netochin
- Life Sciences Center, Moscow Institute of Physics and Technology , Institutsky Per., 9, Dolgoprudny, Moscow Region 141700, Russian Federation
| | - Mikhail M Raihstat
- N. D. Zelinsky Institute of Organic Chemistry, RAS , Leninsky Prospect, 47, 119991, Moscow, Russian Federation
| | - Leonid D Konyushkin
- N. D. Zelinsky Institute of Organic Chemistry, RAS , Leninsky Prospect, 47, 119991, Moscow, Russian Federation
| | - Polina B Volynchuk
- Life Sciences Center, Moscow Institute of Physics and Technology , Institutsky Per., 9, Dolgoprudny, Moscow Region 141700, Russian Federation
| | - Elena I Marusich
- Life Sciences Center, Moscow Institute of Physics and Technology , Institutsky Per., 9, Dolgoprudny, Moscow Region 141700, Russian Federation
| | - Vera V Nazarenko
- Life Sciences Center, Moscow Institute of Physics and Technology , Institutsky Per., 9, Dolgoprudny, Moscow Region 141700, Russian Federation
| | - Sergey V Leonov
- Life Sciences Center, Moscow Institute of Physics and Technology , Institutsky Per., 9, Dolgoprudny, Moscow Region 141700, Russian Federation
- Institute of Cell Biophysics, RAS , Institutskaya Street, 3, Pushchino, Moscow Region 142290, Russian Federation
| | - Alex S Kiselyov
- Life Sciences Center, Moscow Institute of Physics and Technology , Institutsky Per., 9, Dolgoprudny, Moscow Region 141700, Russian Federation
| |
Collapse
|
46
|
Intra-spindle Microtubule Assembly Regulates Clustering of Microtubule-Organizing Centers during Early Mouse Development. Cell Rep 2016; 15:54-60. [PMID: 27052165 DOI: 10.1016/j.celrep.2016.02.087] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/14/2016] [Accepted: 02/24/2016] [Indexed: 11/20/2022] Open
Abstract
Errors during cell division in oocytes and early embryos are linked to birth defects in mammals. Bipolar spindle assembly in early mouse embryos is unique in that three or more acentriolar microtubule-organizing centers (MTOCs) are initially formed and are then clustered into two spindle poles. Using a knockout mouse and live imaging of spindles in embryos, we demonstrate that MTOC clustering during the blastocyst stage requires augmin, a critical complex for MT-dependent MT nucleation within the spindle. Functional analyses in cultured cells with artificially increased numbers of centrosomes indicate that the lack of intra-spindle MT nucleation, but not loss of augmin per se or overall reduction of spindle MTs, is the cause of clustering failure. These data suggest that onset of mitosis with three or more MTOCs is turned into a typical bipolar division through augmin-dependent intra-spindle MT assembly.
Collapse
|