1
|
Monfort-Vengut A, Sanz-Gómez N, Ballesteros-Sánchez S, Ortigosa B, Cambón A, Ramos M, Lorenzo ÁMS, Escribano-Cebrián M, Rosa-Rosa JM, Martínez-López J, Sánchez-Prieto R, Sotillo R, de Cárcer G. Osmotic stress influences microtubule drug response via WNK1 kinase signaling. Drug Resist Updat 2025; 79:101203. [PMID: 39855050 DOI: 10.1016/j.drup.2025.101203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025]
Abstract
Ion homeostasis is critical for numerous cellular processes, and disturbances in ionic balance underlie diverse pathological conditions, including cancer progression. Targeting ion homeostasis is even considered as a strategy to treat cancer. However, very little is known about how ion homeostasis may influence anticancer drug response. In a genome-wide CRISPR-Cas9 resistance drug screen, we identified and validated the master osmostress regulator WNK1 kinase as a modulator of the response to the mitotic inhibitor rigosertib. Osmotic stress and WNK1 inactivation lead to an altered response not only to rigosertib treatment but also to other microtubule-related drugs, minimizing the prototypical mitotic arrest produced by these compounds. This effect is due to an alteration in microtubule stability and polymerization dynamics, likely maintained by fluctuations in intracellular molecular crowding upon WNK1 inactivation. This promotes resistance to microtubule depolymerizing compounds, and increased sensitivity to microtubule stabilizing drugs. In summary, our data proposes WNK1 osmoregulation activity as an important modulator for microtubule-associated chemotherapy response.
Collapse
Affiliation(s)
- Ana Monfort-Vengut
- Cell Cycle & Cancer Biomarkers Laboratory, Cancer Department, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC-UAM, Madrid 28029, Spain
| | - Natalia Sanz-Gómez
- Cell Cycle & Cancer Biomarkers Laboratory, Cancer Department, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC-UAM, Madrid 28029, Spain; Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Sandra Ballesteros-Sánchez
- Cell Cycle & Cancer Biomarkers Laboratory, Cancer Department, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC-UAM, Madrid 28029, Spain; Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Beatriz Ortigosa
- Cell Cycle & Cancer Biomarkers Laboratory, Cancer Department, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC-UAM, Madrid 28029, Spain
| | - Aitana Cambón
- Cell Cycle & Cancer Biomarkers Laboratory, Cancer Department, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC-UAM, Madrid 28029, Spain
| | - Maria Ramos
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Ángela Montes-San Lorenzo
- Cell Cycle & Cancer Biomarkers Laboratory, Cancer Department, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC-UAM, Madrid 28029, Spain; Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - María Escribano-Cebrián
- Cell Cycle & Cancer Biomarkers Laboratory, Cancer Department, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC-UAM, Madrid 28029, Spain; Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Juan Manuel Rosa-Rosa
- Hematology Department, Hospital 12 de Octubre, Madrid 28041, Spain; H12O-CNIO Hematological Tumour Unit, Spanish National Cancer Center (CNIO), Madrid 28029, Spain
| | - Joaquín Martínez-López
- Hematology Department, Hospital 12 de Octubre, Madrid 28041, Spain; H12O-CNIO Hematological Tumour Unit, Spanish National Cancer Center (CNIO), Madrid 28029, Spain; Department of Medicine, Complutense University, Madrid 28040, Spain
| | - Ricardo Sánchez-Prieto
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Molecular Bases of Chemo and Radioresistance in Tumors Laboratory, Cancer Department, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC-UAM, Madrid 28029, Spain; Molecular Oncology Laboratory, Molecular Medicine Unit, Centro Regional de Investigaciones Biomédicas, UCLM, Albacete 02008, Spain; UCLM Biomedicine Unit Associated to CSIC, Spain; CSIC Conexión-Cáncer Hub, Spain
| | - Rocío Sotillo
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Guillermo de Cárcer
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; UCLM Biomedicine Unit Associated to CSIC, Spain; CSIC Conexión-Cáncer Hub, Spain.
| |
Collapse
|
2
|
Gao W, Lu J, Yang Z, Li E, Cao Y, Xie L. Mitotic Functions and Characters of KIF11 in Cancers. Biomolecules 2024; 14:386. [PMID: 38672404 PMCID: PMC11047945 DOI: 10.3390/biom14040386] [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: 02/07/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Mitosis mediates the accurate separation of daughter cells, and abnormalities are closely related to cancer progression. KIF11, a member of the kinesin family, plays a vital role in the formation and maintenance of the mitotic spindle. Recently, an increasing quantity of data have demonstrated the upregulated expression of KIF11 in various cancers, promoting the emergence and progression of cancers. This suggests the great potential of KIF11 as a prognostic biomarker and therapeutic target. However, the molecular mechanisms of KIF11 in cancers have not been systematically summarized. Therefore, we first discuss the functions of the protein encoded by KIF11 during mitosis and connect the abnormal expression of KIF11 with its clinical significance. Then, we elucidate the mechanism of KIF11 to promote various hallmarks of cancers. Finally, we provide an overview of KIF11 inhibitors and outline areas for future work.
Collapse
Affiliation(s)
| | | | | | | | - Yufei Cao
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China; (W.G.); (J.L.); (Z.Y.); (E.L.)
| | - Lei Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China; (W.G.); (J.L.); (Z.Y.); (E.L.)
| |
Collapse
|
3
|
Hunter B, Allingham JS. These motors were made for walking. Protein Sci 2020; 29:1707-1723. [PMID: 32472639 DOI: 10.1002/pro.3895] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/21/2022]
Abstract
Kinesins are a diverse group of adenosine triphosphate (ATP)-dependent motor proteins that transport cargos along microtubules (MTs) and change the organization of MT networks. Shared among all kinesins is a ~40 kDa motor domain that has evolved an impressive assortment of motility and MT remodeling mechanisms as a result of subtle tweaks and edits within its sequence. Several elegant studies of different kinesin isoforms have exposed the purpose of structural changes in the motor domain as it engages and leaves the MT. However, few studies have compared the sequences and MT contacts of these kinesins systematically. Along with clever strategies to trap kinesin-tubulin complexes for X-ray crystallography, new advancements in cryo-electron microscopy have produced a burst of high-resolution structures that show kinesin-MT interfaces more precisely than ever. This review considers the MT interactions of kinesin subfamilies that exhibit significant differences in speed, processivity, and MT remodeling activity. We show how their sequence variations relate to their tubulin footprint and, in turn, how this explains the molecular activities of previously characterized mutants. As more high-resolution structures become available, this type of assessment will quicken the pace toward establishing each kinesin's design-function relationship.
Collapse
Affiliation(s)
- Byron Hunter
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - John S Allingham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
4
|
Suppressor Analysis Uncovers That MAPs and Microtubule Dynamics Balance with the Cut7/Kinesin-5 Motor for Mitotic Spindle Assembly in Schizosaccharomyces pombe. G3-GENES GENOMES GENETICS 2019; 9:269-280. [PMID: 30463883 PMCID: PMC6325904 DOI: 10.1534/g3.118.200896] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The Kinesin-5 motor Cut7 in Schizosaccharomyces pombe plays essential roles in spindle pole separation, leading to the assembly of bipolar spindle. In many organisms, simultaneous inactivation of Kinesin-14s neutralizes Kinesin-5 deficiency. To uncover the molecular network that counteracts Kinesin-5, we have conducted a genetic screening for suppressors that rescue the cut7-22 temperature sensitive mutation, and identified 10 loci. Next generation sequencing analysis reveals that causative mutations are mapped in genes encoding α-, β-tubulins and the microtubule plus-end tracking protein Mal3/EB1, in addition to the components of the Pkl1/Kinesin-14 complex. Moreover, the deletion of various genes required for microtubule nucleation/polymerization also suppresses the cut7 mutant. Intriguingly, Klp2/Kinesin-14 levels on the spindles are significantly increased in cut7 mutants, whereas these increases are negated by suppressors, which may explain the suppression by these mutations/deletions. Consistent with this notion, mild overproduction of Klp2 in these double mutant cells confers temperature sensitivity. Surprisingly, treatment with a microtubule-destabilizing drug not only suppresses cut7 temperature sensitivity but also rescues the lethality resulting from the deletion of cut7, though a single klp2 deletion per se cannot compensate for the loss of Cut7. We propose that microtubule assembly and/or dynamics antagonize Cut7 functions, and that the orchestration between these two factors is crucial for bipolar spindle assembly.
Collapse
|
5
|
Xu X, Wicki-Stordeur LE, Sanchez-Arias JC, Liu M, Weaver MS, Choi CSW, Swayne LA. Probenecid Disrupts a Novel Pannexin 1-Collapsin Response Mediator Protein 2 Interaction and Increases Microtubule Stability. Front Cell Neurosci 2018; 12:124. [PMID: 29867357 PMCID: PMC5958195 DOI: 10.3389/fncel.2018.00124] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/17/2018] [Indexed: 12/25/2022] Open
Abstract
Neurite formation relies on finely-tuned control of the cytoskeleton. Here we identified a novel protein-protein interaction between the ion and metabolite channel protein Pannexin 1 (Panx1) and collapsin response mediator protein 2 (Crmp2), a positive regulator of microtubule polymerization and stabilization. Panx1 and Crmp2 co-precipitated from both Neuro-2a (N2a) cells and mouse ventricular zone (VZ) tissue. In vitro binding assays between purified proteins revealed the interaction occurs directly between the Panx1 C-terminus (Panx1 CT) and Crmp2. Because Crmp2 is a well-established microtubule-stabilizing protein, and we previously observed a marked increase in neurite formation following treatment with the Panx1 blocker, probenecid, in N2a cells and VZ neural precursor cells (NPCs), we investigated the impact of probenecid on the Panx1-Crmp2 interaction. Probenecid treatment significantly disrupted the Panx1-Crmp2 interaction by both immunoprecipitation (IP) and proximity ligation analysis, without altering overall Crmp2 protein expression levels. In the presence of probenecid, Crmp2 was concentrated at the distal ends of growing neurites. Moreover, probenecid treatment increased tubulin polymerization and microtubule stability in N2a cells. These results reveal that probenecid disrupts a novel interaction between Panx1 and the microtubule stabilizer, Crmp2, and also increases microtubule stability.
Collapse
Affiliation(s)
- Xiaoxue Xu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | | | - Mei Liu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nanjing, China
| | - Maria S Weaver
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Catherine S W Choi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Leigh A Swayne
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.,Department of Biology, University of Victoria, Victoria, BC, Canada.,Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Dilworth D, Gudavicius G, Xu X, Boyce AKJ, O’Sullivan C, Serpa JJ, Bilenky M, Petrochenko EV, Borchers CH, Hirst M, Swayne LA, Howard P, Nelson CJ. The prolyl isomerase FKBP25 regulates microtubule polymerization impacting cell cycle progression and genomic stability. Nucleic Acids Res 2018; 46:2459-2478. [PMID: 29361176 PMCID: PMC5861405 DOI: 10.1093/nar/gky008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 12/14/2017] [Accepted: 01/12/2018] [Indexed: 12/25/2022] Open
Abstract
FK506 binding proteins (FKBPs) catalyze the interconversion of cis-trans proline conformers in proteins. Importantly, FK506 drugs have anti-cancer and neuroprotective properties, but the effectors and mechanisms underpinning these properties are not well understood because the cellular function(s) of most FKBP proteins are unclear. FKBP25 is a nuclear prolyl isomerase that interacts directly with nucleic acids and is associated with several DNA/RNA binding proteins. Here, we show the catalytic FKBP domain binds microtubules (MTs) directly to promote their polymerization and stabilize the MT network. Furthermore, FKBP25 associates with the mitotic spindle and regulates entry into mitosis. This interaction is important for mitotic spindle dynamics, as we observe increased chromosome instability in FKBP25 knockdown cells. Finally, we provide evidence that FKBP25 association with chromatin is cell-cycle regulated by Protein Kinase C phosphorylation. This disrupts FKBP25-DNA contacts during mitosis while maintaining its interaction with the spindle apparatus. Collectively, these data support a model where FKBP25 association with chromatin and MTs is carefully choreographed to ensure faithful genome duplication. Additionally, they highlight that FKBP25 is a MT-associated FK506 receptor and potential therapeutic target in MT-associated diseases.
Collapse
Affiliation(s)
- David Dilworth
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Geoff Gudavicius
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Xiaoxue Xu
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Andrew K J Boyce
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Connor O’Sullivan
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Jason J Serpa
- University of Victoria Genome BC Proteomics Centre, Vancouver Island Technology Park, Victoria, BC, V8Z 7X8, Canada
| | - Misha Bilenky
- BC Cancer Agency Genome Sciences Centre and the Department of Microbiology & Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Evgeniy V Petrochenko
- University of Victoria Genome BC Proteomics Centre, Vancouver Island Technology Park, Victoria, BC, V8Z 7X8, Canada
| | - Christoph H Borchers
- University of Victoria Genome BC Proteomics Centre, Vancouver Island Technology Park, Victoria, BC, V8Z 7X8, Canada
| | - Martin Hirst
- BC Cancer Agency Genome Sciences Centre and the Department of Microbiology & Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Perry Howard
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Christopher J Nelson
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| |
Collapse
|
7
|
Pei YY, Li GC, Ran J, Wei FX. Kinesin family member 11 contributes to the progression and prognosis of human breast cancer. Oncol Lett 2017; 14:6618-6626. [PMID: 29181100 PMCID: PMC5696720 DOI: 10.3892/ol.2017.7053] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 06/02/2017] [Indexed: 01/24/2023] Open
Abstract
The present study aimed to clarify the association between kinesin family member 11 (KIF11) and human breast cancer, and the effect of KIF11 on breast cancer cell progression. Western blot analysis, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis, retroviral infection, immunohistochemistry staining, MTT assay, anchorage-independent growth ability assay and tumorigenicity assay were all used in the present study. Western blot and RT-qPCR analysis revealed that the expression of KIF11 was markedly increased in malignant cells compared with that in non-tumorous cells at the mRNA and protein level. Immunohistochemical analysis revealed that KIF11 expression was upregulated in 256/268 (95.8%) paraffin-embedded archival breast cancer biopsies. Statistical analysis demonstrated a significant association between the upregulation of KIF11 expression and the progression of breast cancer. Multivariate analysis revealed that KIF11 upregulation represents an independent prognostic indicator for the survival of patients with breast cancer. Tumorigenicity experiments were further used to evaluate the effect of KIF11 in non-obese diabetic/severe combined immunodeficient mice. Silencing endogenous KIF11 by short hairpin RNAs inhibited the proliferation of breast cancer cells in vitro and in vivo. The present results suggest that KIF11 may serve an important function in the proliferation of breast cancer and may represent a novel and useful prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Yuan-Yuan Pei
- Shenzhen Longgang Maternal and Child Health Hospital Centralab, Shenzhen, Guangdong 518172, P.R. China
| | - Gao-Chi Li
- Shenzhen Longgang Maternal and Child Health Hospital Centralab, Shenzhen, Guangdong 518172, P.R. China
| | - Jian Ran
- Shenzhen Longgang Maternal and Child Health Hospital Centralab, Shenzhen, Guangdong 518172, P.R. China
| | - Feng-Xiang Wei
- Shenzhen Longgang Maternal and Child Health Hospital Centralab, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
8
|
Gaume X, Place C, Delage H, Mongelard F, Monier K, Bouvet P. Expression of Nucleolin Affects Microtubule Dynamics. PLoS One 2016; 11:e0157534. [PMID: 27309529 PMCID: PMC4911123 DOI: 10.1371/journal.pone.0157534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 06/01/2016] [Indexed: 02/01/2023] Open
Abstract
Nucleolin is present in diverse cellular compartments and is involved in a variety of cellular processes from nucleolar structure and function to intracellular trafficking, cell adhesion and migration. Recently, nucleolin has been localized at the mature centriole where it is involved in microtubule nucleation and anchoring. Although this new function of nucleolin linked to microtubule regulation has been identified, the global effects of nucleolin on microtubule dynamics have not been addressed yet. In the present study, we analyzed the roles of nucleolin protein levels on global microtubule dynamics by tracking the EB3 microtubule plus end binding protein in live cells. We have found that during microtubule growth phases, nucleolin affects both the speed and life time of polymerization and by analyzing catastrophe events, we showed that nucleolin reduces catastrophe frequency. This new property of nucleolin was then confirmed in a cold induced microtubule depolymerization experiment in which we have found that cold resistant microtubules were totally destabilized in nucleolin depleted cells. Altogether, our data demonstrate a new function of nucleolin on microtubule stabilization, thus bringing novel insights into understanding the multifunctional properties of nucleolin in healthy and cancer cells.
Collapse
Affiliation(s)
- Xavier Gaume
- Université de Lyon, Ecole normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Christophe Place
- Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard, CNRS, Laboratoire de Physique, F-69342, Lyon, France
| | - Helene Delage
- Université de Lyon, Ecole normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Fabien Mongelard
- Université de Lyon, Ecole normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Karine Monier
- Université de Lyon, Ecole normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Philippe Bouvet
- Université de Lyon, Ecole normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| |
Collapse
|
9
|
Abstract
Kinesin-5 slides antiparallel microtubules during spindle assembly, and regulates the branching of growing axons. Besides the mechanical activities enabled by its tetrameric configuration, the specific motor properties of kinesin-5 that underlie its cellular function remain unclear. Here by engineering a stable kinesin-5 dimer and reconstituting microtubule dynamics in vitro, we demonstrate that kinesin-5 promotes microtubule polymerization by increasing the growth rate and decreasing the catastrophe frequency. Strikingly, microtubules growing in the presence of kinesin-5 have curved plus ends, suggesting that the motor stabilizes growing protofilaments. Single-molecule fluorescence experiments reveal that kinesin-5 remains bound to the plus ends of static microtubules for 7 s, and tracks growing microtubule plus ends in a manner dependent on its processivity. We propose that kinesin-5 pauses at microtubule plus ends and enhances polymerization by stabilizing longitudinal tubulin-tubulin interactions, and that these activities underlie the ability kinesin-5 to slide and stabilize microtubule bundles in cells.
Collapse
|
10
|
Larrieu D, Britton S, Demir M, Rodriguez R, Jackson SP. Chemical inhibition of NAT10 corrects defects of laminopathic cells. Science 2014; 344:527-32. [PMID: 24786082 PMCID: PMC4246063 DOI: 10.1126/science.1252651] [Citation(s) in RCA: 282] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Down-regulation and mutations of the nuclear-architecture proteins lamin A and C cause misshapen nuclei and altered chromatin organization associated with cancer and laminopathies, including the premature-aging disease Hutchinson-Gilford progeria syndrome (HGPS). Here, we identified the small molecule "Remodelin" that improved nuclear architecture, chromatin organization, and fitness of both human lamin A/C-depleted cells and HGPS-derived patient cells and decreased markers of DNA damage in these cells. Using a combination of chemical, cellular, and genetic approaches, we identified the acetyl-transferase protein NAT10 as the target of Remodelin that mediated nuclear shape rescue in laminopathic cells via microtubule reorganization. These findings provide insights into how NAT10 affects nuclear architecture and suggest alternative strategies for treating laminopathies and aging.
Collapse
Affiliation(s)
- Delphine Larrieu
- The Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, United Kingdom
| | - Sébastien Britton
- The Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, United Kingdom
| | - Mukerrem Demir
- The Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, United Kingdom
| | - Raphaël Rodriguez
- Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Stephen P. Jackson
- The Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, United Kingdom
- The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| |
Collapse
|
11
|
Kinesin-5: cross-bridging mechanism to targeted clinical therapy. Gene 2013; 531:133-49. [PMID: 23954229 DOI: 10.1016/j.gene.2013.08.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/29/2013] [Accepted: 08/02/2013] [Indexed: 12/28/2022]
Abstract
Kinesin motor proteins comprise an ATPase superfamily that works hand in hand with microtubules in every eukaryote. The mitotic kinesins, by virtue of their potential therapeutic role in cancerous cells, have been a major focus of research for the past 28 years since the discovery of the canonical Kinesin-1 heavy chain. Perhaps the simplest player in mitotic spindle assembly, Kinesin-5 (also known as Kif11, Eg5, or kinesin spindle protein, KSP) is a plus-end-directed motor localized to interpolar spindle microtubules and to the spindle poles. Comprised of a homotetramer complex, its function primarily is to slide anti-parallel microtubules apart from one another. Based on multi-faceted analyses of this motor from numerous laboratories over the years, we have learned a great deal about the function of this motor at the atomic level for catalysis and as an integrated element of the cytoskeleton. These data have, in turn, informed the function of motile kinesins on the whole, as well as spearheaded integrative models of the mitotic apparatus in particular and regulation of the microtubule cytoskeleton in general. We review what is known about how this nanomotor works, its place inside the cytoskeleton of cells, and its small-molecule inhibitors that provide a toolbox for understanding motor function and for anticancer treatment in the clinic.
Collapse
|
12
|
Tan LJ, Saijo M, Kuraoka I, Narita T, Takahata C, Iwai S, Tanaka K. Xeroderma pigmentosum group F protein binds to Eg5 and is required for proper mitosis: implications for XP-F and XFE. Genes Cells 2012; 17:173-85. [PMID: 22353549 DOI: 10.1111/j.1365-2443.2012.01582.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The xeroderma pigmentosum group F-cross-complementing rodent repair deficiency group 1 (XPF-ERCC1) complex is a structure-specific endonuclease involved in nucleotide excision repair (NER) and interstrand cross-link (ICL) repair. Patients with XPF mutations may suffer from two forms of xeroderma pigmentosum (XP): XP-F patients show mild photosensitivity and proneness to skin cancer but rarely show any neurological abnormalities, whereas XFE patients display symptoms of severe XP symptoms, growth retardation and accelerated aging. Xpf knockout mice display accelerated aging and die before weaning. These results suggest that the XPF-ERCC1 complex has additional functions besides NER and ICL repair and is essential for development and growth. In this study, we show a partial colocalization of XPF with mitotic spindles and Eg5. XPF knockdown in cells led to an increase in the frequency of abnormal nuclear morphology and mitosis. Similarly, the frequency of abnormal nuclei and mitosis was increased in XP-F and XFE cells. In addition, we showed that Eg5 enhances the action of XPF-ERCC1 nuclease activity. Taken together, these results suggest that the interaction between XPF and Eg5 plays a role in mitosis and DNA repair and offer new insights into the pathogenesis of XP-F and XFE.
Collapse
Affiliation(s)
- Li Jing Tan
- Human Cell Biology Group, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Drummond DR. Regulation of microtubule dynamics by kinesins. Semin Cell Dev Biol 2011; 22:927-34. [PMID: 22001250 DOI: 10.1016/j.semcdb.2011.09.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 09/30/2011] [Indexed: 01/14/2023]
Abstract
The simple mechanistic and functional division of the kinesin family into either active translocators or non-motile microtubule depolymerases was initially appropriate but is now proving increasingly unhelpful, given evidence that several translocase kinesins can affect microtubule dynamics, whilst non-translocase kinesins can promote microtubule assembly and depolymerisation. Such multi-role kinesins act either directly on microtubule dynamics, by interaction with microtubules and tubulin, or indirectly, through the transport of other factors along the lattice to the microtubule tip. Here I review recent progress on the mechanisms and roles of these translocase kinesins.
Collapse
Affiliation(s)
- Douglas R Drummond
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| |
Collapse
|
14
|
Abstract
M-CSF-driven differentiation of peripheral blood monocytes is one of the sources of tissue macrophages. In humans and mice, the differentiation process involves the activation of caspases that cleave a limited number of proteins. One of these proteins is nucleophosmin (NPM1), a multifunctional and ubiquitous protein. Here, we show that caspases activated in monocytes exposed to M-CSF cleave NPM1 at D213 to generate a 30-kDa N-terminal fragment. The protein is further cleaved into a 20-kDa fragment, which involves cathepsin B. NPM1 fragments contribute to the limited motility, migration, and phagocytosis capabilities of resting macrophages. Their activation with lipopolysaccharides inhibits proteolytic processes and restores expression of the full-length protein that negatively regulates the transcription of genes encoding inflammatory cytokines (eg, NPM1 is recruited with NF-κB on the MCP1 gene promoter to decrease its transcription). In mice with heterozygous npm gene deletion, cytokine production in response to lipopolysaccharides, including CXCL1 (KC), MCP1, and MIP2, is dramatically enhanced. These results indicate a dual function of NPM1 in M-CSF-differentiated macrophages. Proteolysis of the protein participates in the establishment of a mature macrophage phenotype. In response to inflammatory stimuli, the full-length protein negatively regulates inflammatory cytokine production.
Collapse
|
15
|
Daire V, Poüs C. Kinesins and protein kinases: key players in the regulation of microtubule dynamics and organization. Arch Biochem Biophys 2011; 510:83-92. [PMID: 21345331 DOI: 10.1016/j.abb.2011.02.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 02/05/2011] [Accepted: 02/11/2011] [Indexed: 02/04/2023]
Abstract
Microtubule dynamics is controlled and amplified in vivo by complex sets of regulators. Among these regulatory proteins, molecular motors from the kinesin superfamily are taking an increasing importance. Here we review how microtubule disassembly or assembly into interphase microtubules, mitotic spindle or cilia may involve kinesins and how protein kinases may participate in these kinesin-dependent regulations.
Collapse
Affiliation(s)
- Vanessa Daire
- UPRES EA, Univ. Paris-Sud, Faculté de Pharmacie, Châtenay-Malabry, France
| | | |
Collapse
|
16
|
NPM1/B23: A Multifunctional Chaperone in Ribosome Biogenesis and Chromatin Remodeling. Biochem Res Int 2010; 2011:195209. [PMID: 21152184 PMCID: PMC2989734 DOI: 10.1155/2011/195209] [Citation(s) in RCA: 227] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 08/29/2010] [Indexed: 12/21/2022] Open
Abstract
At a first glance, ribosome biogenesis and chromatin remodeling are quite different processes, but they share a common problem involving interactions between charged nucleic acids and small basic proteins that may result in unwanted intracellular aggregations. The multifunctional nuclear acidic chaperone NPM1 (B23/nucleophosmin) is active in several stages of ribosome biogenesis, chromatin remodeling, and mitosis as well as in DNA repair, replication and transcription. In addition, NPM1 plays an important role in the Myc-ARF-p53 pathway as well as in SUMO regulation. However, the relative importance of NPM1 in these processes remains unclear. Provided herein is an update on the expanding list of the diverse activities and interacting partners of NPM1. Mechanisms of NPM1 nuclear export functions of NPM1 in the nucleolus and at the mitotic spindle are discussed in relation to tumor development. It is argued that the suggested function of NPM1 as a histone chaperone could explain several, but not all, of the effects observed in cells following changes in NPM1 expression. A future challenge is to understand how NPM1 is activated, recruited, and controlled to carry out its functions.
Collapse
|