151
|
Targeting of Fzr/Cdh1 for timely activation of the APC/C at the centrosome during mitotic exit. Nat Commun 2016; 7:12607. [PMID: 27558644 PMCID: PMC5007356 DOI: 10.1038/ncomms12607] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 07/16/2016] [Indexed: 01/06/2023] Open
Abstract
A multi-subunit ubiquitin ligase, the anaphase-promoting complex/cyclosome (APC/C), regulates critical cellular processes including the cell cycle. To accomplish its diverse functions, APC/C activity must be precisely regulated in time and space. The interphase APC/C activator Fizzy-related (Fzr or Cdh1) is localized at centrosomes in animal cells. However, neither the mechanism of its localization nor its importance is clear. Here we identify the centrosome component Spd2 as a major partner of Fzr in Drosophila. The localization of Fzr to the centriole during interphase depends on direct interaction with Spd2. By generating Spd2 mutants unable to bind Fzr, we show that centrosomal localization of Fzr is essential for optimal APC/C activation towards its centrosomal substrate Aurora A. Finally, we show that Spd2 is also a novel APC/CFzr substrate. Our study is the first to demonstrate the critical importance of distinct subcellular pools of APC/C activators in the spatiotemporal control of APC/C activity. The activity of the anaphase-promoting complex/cyclosome (APC/C) needs to be regulated in time and space to perform different functions. Here the authors show that Spd2 localizes the APC/C activator Fzr at the centrosomes to promote optimal APC/C activity towards its centrosomal substrate Aurora A.
Collapse
|
152
|
Yang X, Xu R, Lin Y, Zhen Y, Wei J, Hu G, Sun H. Recombinant adenovirus of human p66Shc inhibits MCF-7 cell proliferation. Sci Rep 2016; 6:31534. [PMID: 27530145 PMCID: PMC4987618 DOI: 10.1038/srep31534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/21/2016] [Indexed: 01/03/2023] Open
Abstract
The aim of this work was to construct a human recombinant p66Shc adenovirus and to investigate the inhibition of recombinant p66Shc adenovirus on MCF-7 cells. The recombinant adenovirus expression vector was constructed using the Adeno-X Adenoviral System 3. Inhibition of MCF-7 cell proliferation was determined by MTT. Intracellular ROS was measured by DCFH-DA fluorescent probes, and 8-OHdG was detected by ELISA. Cell apoptosis and the cell cycle were assayed by flow cytometry. Western blot were used to observe protein expression. p66Shc expression was upregulated in 4 cell lines after infection. The inhibitory effect of p66Shc recombinant adenovirus on MCF-7 cells was accompanied by enhanced ROS and 8-OHdG. However, no significant differences were observed in the cell apoptosis rate. The ratio of the cell cycle G2/M phase showed a significant increase. Follow-up experiments demonstrated that the expressions of p53, p-p53, cyclin B1 and CDK1 were upregulated with the overexpression of p66Shc. The Adeno-X Adenoviral System 3 can be used to efficiently construct recombinant adenovirus containing p66Shc gene, and the Adeno-X can inhibit the proliferation of MCF-7 cells by inducing cell cycle arrest at the G2/M phase. These results suggested that p66Shc may be a key target for clinical cancer therapy.
Collapse
Affiliation(s)
- Xiaoshan Yang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Peking Union Medical College &Chinese Academy of Medical Sciences, Tianjin 300192, China.,The key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing, 100730, China
| | - Rong Xu
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Peking Union Medical College &Chinese Academy of Medical Sciences, Tianjin 300192, China.,The key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing, 100730, China
| | - Yajun Lin
- The key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing, 100730, China
| | - Yongzhan Zhen
- Department of Histology and Embryology, College of Basic Medical, Hebei United University, Tangshan, 063000, China
| | - Jie Wei
- The key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing, 100730, China
| | - Gang Hu
- The key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing, 100730, China
| | - Hongfan Sun
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Peking Union Medical College &Chinese Academy of Medical Sciences, Tianjin 300192, China
| |
Collapse
|
153
|
Al Nakouzi N, Wang CK, Beraldi E, Jager W, Ettinger S, Fazli L, Nappi L, Bishop J, Zhang F, Chauchereau A, Loriot Y, Gleave M. Clusterin knockdown sensitizes prostate cancer cells to taxane by modulating mitosis. EMBO Mol Med 2016; 8:761-78. [PMID: 27198502 PMCID: PMC4931290 DOI: 10.15252/emmm.201506059] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Clusterin (CLU) is a stress‐activated molecular chaperone that confers treatment resistance to taxanes when highly expressed. While CLU inhibition potentiates activity of taxanes and other anti‐cancer therapies in preclinical models, progression to treatment‐resistant disease still occurs implicating additional compensatory survival mechanisms. Taxanes are believed to selectively target cells in mitosis, a complex mechanism controlled in part by balancing antagonistic roles of Cdc25C and Wee1 in mitosis progression. Our data indicate that CLU silencing induces a constitutive activation of Cdc25C, which delays mitotic exit and hence sensitizes cancer cells to mitotic‐targeting agents such as taxanes. Unchecked Cdc25C activation leads to mitotic catastrophe and cell death unless cells up‐regulate protective mechanisms mediated through the cell cycle regulators Wee1 and Cdk1. In this study, we show that CLU silencing induces a constitutive activation of Cdc25C via the phosphatase PP2A leading to relief of negative feedback inhibition and activation of Wee1‐Cdk1 to promote survival and limit therapeutic efficacy. Simultaneous inhibition of CLU‐regulated cell cycle effector Wee1 may improve synergistic responses of biologically rational combinatorial regimens using taxanes and CLU inhibitors.
Collapse
Affiliation(s)
- Nader Al Nakouzi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Chris Kedong Wang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eliana Beraldi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Wolfgang Jager
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Susan Ettinger
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ladan Fazli
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lucia Nappi
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer Bishop
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Fan Zhang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Anne Chauchereau
- Department of Cancer Medicine, Gustave Roussy, Cancer Campus, Grand Paris, University of Paris-Sud, Villejuif, France INSERM U981, Villejuif, France
| | - Yohann Loriot
- Department of Cancer Medicine, Gustave Roussy, Cancer Campus, Grand Paris, University of Paris-Sud, Villejuif, France INSERM U981, Villejuif, France
| | - Martin Gleave
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
154
|
Klochendler A, Caspi I, Corem N, Moran M, Friedlich O, Elgavish S, Nevo Y, Helman A, Glaser B, Eden A, Itzkovitz S, Dor Y. The Genetic Program of Pancreatic β-Cell Replication In Vivo. Diabetes 2016; 65:2081-93. [PMID: 26993067 PMCID: PMC4915587 DOI: 10.2337/db16-0003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 01/01/2023]
Abstract
The molecular program underlying infrequent replication of pancreatic β-cells remains largely inaccessible. Using transgenic mice expressing green fluorescent protein in cycling cells, we sorted live, replicating β-cells and determined their transcriptome. Replicating β-cells upregulate hundreds of proliferation-related genes, along with many novel putative cell cycle components. Strikingly, genes involved in β-cell functions, namely, glucose sensing and insulin secretion, were repressed. Further studies using single-molecule RNA in situ hybridization revealed that in fact, replicating β-cells double the amount of RNA for most genes, but this upregulation excludes genes involved in β-cell function. These data suggest that the quiescence-proliferation transition involves global amplification of gene expression, except for a subset of tissue-specific genes, which are "left behind" and whose relative mRNA amount decreases. Our work provides a unique resource for the study of replicating β-cells in vivo.
Collapse
Affiliation(s)
- Agnes Klochendler
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Inbal Caspi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Corem
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Maya Moran
- Department of Cell and Developmental Biology, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oriel Friedlich
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Sharona Elgavish
- Info-CORE, Bioinformatics Unit of the I-CORE Computation Center, and Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yuval Nevo
- Info-CORE, Bioinformatics Unit of the I-CORE Computation Center, and Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Aharon Helman
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Benjamin Glaser
- Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amir Eden
- Department of Cell and Developmental Biology, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
155
|
Magidson V, He J, Ault JG, O'Connell CB, Yang N, Tikhonenko I, McEwen BF, Sui H, Khodjakov A. Unattached kinetochores rather than intrakinetochore tension arrest mitosis in taxol-treated cells. J Cell Biol 2016; 212:307-19. [PMID: 26833787 PMCID: PMC4748573 DOI: 10.1083/jcb.201412139] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Taxol induces extensive structural reorganization of the mammalian kinetochore; however, this reorganization is not sufficient to maintain a long-term mitotic arrest unless some of the kinetochores completely lose their attachment to microtubules. Kinetochores attach chromosomes to the spindle microtubules and signal the spindle assembly checkpoint to delay mitotic exit until all chromosomes are attached. Light microscopy approaches aimed to indirectly determine distances between various proteins within the kinetochore (termed Delta) suggest that kinetochores become stretched by spindle forces and compact elastically when the force is suppressed. Low Delta is believed to arrest mitotic progression in taxol-treated cells. However, the structural basis of Delta remains unknown. By integrating same-kinetochore light microscopy and electron microscopy, we demonstrate that the value of Delta is affected by the variability in the shape and size of outer kinetochore domains. The outer kinetochore compacts when spindle forces are maximal during metaphase. When the forces are weakened by taxol treatment, the outer kinetochore expands radially and some kinetochores completely lose microtubule attachment, a condition known to arrest mitotic progression. These observations offer an alternative interpretation of intrakinetochore tension and question whether Delta plays a direct role in the control of mitotic progression.
Collapse
Affiliation(s)
- Valentin Magidson
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - Jie He
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - Jeffrey G Ault
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | | | - Nachen Yang
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - Irina Tikhonenko
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - Bruce F McEwen
- Wadsworth Center, New York State Department of Health, Albany, NY 12201 School of Public Health, State University of New York, Albany, NY 12201
| | - Haixin Sui
- Wadsworth Center, New York State Department of Health, Albany, NY 12201 School of Public Health, State University of New York, Albany, NY 12201
| | - Alexey Khodjakov
- Wadsworth Center, New York State Department of Health, Albany, NY 12201 Rensselaer Polytechnic Institute, Troy, NY 12180
| |
Collapse
|
156
|
Vora SM, Phillips BT. The benefits of local depletion: The centrosome as a scaffold for ubiquitin-proteasome-mediated degradation. Cell Cycle 2016; 15:2124-2134. [PMID: 27294844 DOI: 10.1080/15384101.2016.1196306] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The centrosome is the major microtubule-organizing center in animal cells but is dispensable for proper microtubule spindle formation in many biological contexts and is thus thought to fulfill additional functions. Recent observations suggest that the centrosome acts as a scaffold for proteasomal degradation in the cell to regulate a variety of biological processes including cell fate acquisition, cell cycle control, stress response, and cell morphogenesis. Here, we review the body of studies indicating a role for the centrosome in promoting proteasomal degradation of ubiquitin-proteasome substrates and explore the functional relevance of this system in different biological contexts. We discuss a potential role for the centrosome in coordinating local degradation of proteasomal substrates, allowing cells to achieve stringent spatiotemporal control over various signaling processes.
Collapse
Affiliation(s)
- Setu M Vora
- a Department of Biological Sciences, University of Iowa , Iowa City , IA , USA
| | - Bryan T Phillips
- a Department of Biological Sciences, University of Iowa , Iowa City , IA , USA
| |
Collapse
|
157
|
Höckner S, Neumann-Arnold L, Seufert W. Dual control by Cdk1 phosphorylation of the budding yeast APC/C ubiquitin ligase activator Cdh1. Mol Biol Cell 2016; 27:2198-212. [PMID: 27226481 PMCID: PMC4945139 DOI: 10.1091/mbc.e15-11-0787] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/18/2016] [Indexed: 12/15/2022] Open
Abstract
The antagonism between cyclin-dependent kinases (Cdks) and the ubiquitin ligase APC/C-Cdh1 is central to eukaryotic cell cycle control. APC/C-Cdh1 targets cyclin B and other regulatory proteins for degradation, whereas Cdks disable APC/C-Cdh1 through phosphorylation of the Cdh1 activator protein at multiple sites. Budding yeast Cdh1 carries nine Cdk phosphorylation sites in its N-terminal regulatory domain, most or all of which contribute to inhibition. However, the precise role of individual sites has remained unclear. Here, we report that the Cdk phosphorylation sites of yeast Cdh1 are organized into autonomous subgroups and act through separate mechanisms. Cdk sites 1-3 had no direct effect on the APC/C binding of Cdh1 but inactivated a bipartite nuclear localization sequence (NLS) and thereby controlled the partitioning of Cdh1 between cytoplasm and nucleus. In contrast, Cdk sites 4-9 did not influence the cell cycle-regulated localization of Cdh1 but prevented its binding to the APC/C. Cdk sites 4-9 reside near two recently identified APC/C interaction motifs in a pattern conserved with the human Cdh1 orthologue. Thus a Cdk-inhibited NLS goes along with Cdk-inhibited APC/C binding sites in yeast Cdh1 to relay the negative control by Cdk1 phosphorylation of the ubiquitin ligase APC/C-Cdh1.
Collapse
Affiliation(s)
- Sebastian Höckner
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| | - Lea Neumann-Arnold
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| | - Wolfgang Seufert
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| |
Collapse
|
158
|
Weiderhold KN, Fadri-Moskwik M, Pan J, Nishino M, Chuang C, Deeraksa A, Lin SH, Yu-Lee LY. Dynamic Phosphorylation of NudC by Aurora B in Cytokinesis. PLoS One 2016; 11:e0153455. [PMID: 27074040 PMCID: PMC4830538 DOI: 10.1371/journal.pone.0153455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/30/2016] [Indexed: 01/06/2023] Open
Abstract
Nuclear distribution protein C (NudC) is a mitotic regulator that plays a role in cytokinesis. However, how NudC is regulated during cytokinesis remains unclear. Here, we show that NudC is phosphorylated by Aurora B, a kinase critical for cell abscission. NudC is co-localized with Aurora B at the midbody and co-immunoprecipitated with Aurora B in mitosis. Inhibition of Aurora B by ZM447439 reduced NudC phosphorylation, suggesting that NudC is an Aurora B substrate in vivo. We identified T40 on NudC as an Aurora B phosphorylation site. NudC depletion resulted in cytokinesis failure with a dramatic elongation of the intercellular bridge between daughter cells, sustained Aurora B activity at the midbody, and reduced cell abscission. These cytokinetic defects can be rescued by the ectopic expression of wild-type NudC. Reconstitution with T40A phospho-defective NudC was found to rescue the cytokinesis defect. In contrast, reconstitution with the T40D phospho-mimetic NudC was inefficient in supporting the completion of cytokinesis. These results suggest that that dynamic phosphorylation of NudC by Aurora B regulates cytokinesis.
Collapse
Affiliation(s)
- Kimberly N. Weiderhold
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States of America
| | - Maria Fadri-Moskwik
- Department of Medicine, Section of Allergy Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jing Pan
- Department of Medicine, Section of Allergy Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Michiya Nishino
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States of America
| | - Carol Chuang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Arpaporn Deeraksa
- Department of Medicine, Section of Allergy Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Li-Yuan Yu-Lee
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Medicine, Section of Allergy Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
159
|
Interphase APC/C-Cdc20 inhibition by cyclin A2-Cdk2 ensures efficient mitotic entry. Nat Commun 2016; 7:10975. [PMID: 26960431 PMCID: PMC4792957 DOI: 10.1038/ncomms10975] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/08/2016] [Indexed: 01/19/2023] Open
Abstract
Proper cell-cycle progression requires tight temporal control of the Anaphase Promoting Complex/Cyclosome (APC/C), a large ubiquitin ligase that is activated by one of two co-activators, Cdh1 or Cdc20. APC/C and Cdc20 are already present during interphase but APC/C-Cdc20 regulation during this window of the cell cycle, if any, is unknown. Here we show that cyclin A2-Cdk2 binds and phosphorylates Cdc20 in interphase and this inhibits APC/C-Cdc20 activity. Preventing Cdc20 phosphorylation results in pre-mature activation of the APC/C-Cdc20 and several substrates, including cyclin B1 and A2, are destabilized which lengthens G2 and slows mitotic entry. Expressing non-degradable cyclin A2 but not cyclin B1 restores mitotic entry in these cells. We have thus uncovered a novel positive feedback loop centred on cyclin A2-Cdk2 inhibition of interphase APC/C-Cdc20 to allow further cyclin A2 accumulation and mitotic entry.
Collapse
|
160
|
Expression and Clinical Role of Cdc5L as a Novel Cell Cycle Protein in Hepatocellular Carcinoma. Dig Dis Sci 2016; 61:795-805. [PMID: 26553251 DOI: 10.1007/s10620-015-3937-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 10/22/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Cell division cycle 5-like (Cdc5L), as a pre-mRNA splicing factor, is a regulator of mitotic progression. Previous study found that deletion of endogenous Cdc5L decreases the cell viability via dramatic mitotic arrest, while the role of Cdc5L in cancer biology remains under debate. AIMS To investigate the involvement of Cdc5L in the progression of hepatocellular carcinoma (HCC). METHODS In this study, the expression of Cdc5L was evaluated by Western blot in 8 paired fresh HCC tissues and immunohistochemistry on 116 paraffin-embedded slices. We treated HCC cells by nocodazole to analyze the role of Cdc5L in mitotic progress. To determine whether Cdc5L could regulate the proliferation of HCC cells, we increased endogenous Cdc5L and analyzed the proliferation of HCC cells using Western blot, CCK8, flow cytometry assays, and colony formation analyses. Furthermore, Cdc5L-siRNA oligos were used to confirm that Cdc5L plays an essential role in HCC development. RESULTS Cdc5L was highly expressed in HCC and significantly associated with multiple clinicopathological factors, including AJCC stage, tumor size, and Ki-67. Besides, univariate and multivariate survival analyses demonstrated that high Cdc5L expression was an independent prognostic factor for HCC patients' poor survival. Overexpression of Cdc5L favors cell cycle progress of HCC cells, while downregulation of Cdc5L results in cell cycle arrest at G2/M phase and reduced cell proliferation of HCC cells. CONCLUSIONS Our findings suggested that Cdc5L could play an important role in the tumorigenesis of HCC and thus be a potential therapeutical target to prevent HCC progression.
Collapse
|
161
|
Boekhout M, Yuan R, Wondergem AP, Segeren HA, van Liere EA, Awol N, Jansen I, Wolthuis RMF, de Bruin A, Westendorp B. Feedback regulation between atypical E2Fs and APC/CCdh1 coordinates cell cycle progression. EMBO Rep 2016; 17:414-27. [PMID: 26882548 DOI: 10.15252/embr.201540984] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 01/07/2016] [Indexed: 01/20/2023] Open
Abstract
E2F transcription factors control the oscillating expression pattern of multiple target genes during the cell cycle. Activator E2Fs, E2F1-3, induce an upswing of E2F targets, which is essential for the G1-to-S phase transition, whereas atypical E2Fs, E2F7 and E2F8, mediate a downswing of the same targets during late S, G2, and M phases. Expression of atypical E2Fs is induced by E2F1-3, but it is unknown how atypical E2Fs are inactivated in a timely manner. Here, we demonstrate that E2F7 and E2F8 are substrates of the anaphase-promoting complex/cyclosome (APC/C). Removal of CDH1, or mutating the CDH1-interacting KEN boxes, stabilized E2F7/8 from anaphase onwards and during G1. Expressing KEN mutant E2F7 during G1 impairs S phase entry and eventually results in cell death. Furthermore, we show that E2F8, but not E2F7, interacts also with APC/C(C) (dc20). Importantly, atypical E2Fs can activate APC/C(C) (dh1) by repressing its inhibitors cyclin A, cyclin E, and Emi1. In conclusion, we discovered a feedback loop between atypical E2Fs and APC/C(C) (dh1), which ensures balanced expression of cell cycle genes and normal cell cycle progression.
Collapse
Affiliation(s)
- Michiel Boekhout
- Division of Cell Biology I (B5), The Netherlands Cancer Institute (NKI-AvL), Amsterdam, The Netherlands
| | - Ruixue Yuan
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Annelotte P Wondergem
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Hendrika A Segeren
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Elsbeth A van Liere
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Nesibu Awol
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Imke Jansen
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Rob M F Wolthuis
- Division of Cell Biology I (B5), The Netherlands Cancer Institute (NKI-AvL), Amsterdam, The Netherlands
| | - Alain de Bruin
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands Department of Pediatrics, Division of Molecular Genetics, University Medical Center Groningen University of Groningen, Groningen, The Netherlands
| | - Bart Westendorp
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
162
|
Caous R, Pascal A, Romé P, Richard-Parpaillon L, Karess R, Giet R. Spindle assembly checkpoint inactivation fails to suppress neuroblast tumour formation in aurA mutant Drosophila. Nat Commun 2015; 6:8879. [PMID: 26568519 PMCID: PMC4660220 DOI: 10.1038/ncomms9879] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 10/10/2015] [Indexed: 12/18/2022] Open
Abstract
Tissue homeostasis requires accurate control of cell proliferation, differentiation and chromosome segregation. Drosophila sas-4 and aurA mutants present brain tumours with extra neuroblasts (NBs), defective mitotic spindle assembly and delayed mitosis due to activation of the spindle assembly checkpoint (SAC). Here we inactivate the SAC in aurA and sas-4 mutants to determine whether the generation of aneuploidy compromises NB proliferation. Inactivation of the SAC in the sas-4 mutant impairs NB proliferation and disrupts euploidy. By contrast, disrupting the SAC in the aurA mutant does not prevent NB amplification, tumour formation or chromosome segregation. The monitoring of Mad2 and cyclin B dynamics in live aurA NBs reveals that SAC satisfaction is not coupled to cyclin B degradation. Thus, the NBs of aurA mutants present delayed mitosis, with accurate chromosome segregation occurring in a SAC-independent manner. We report here the existence of an Aurora A-dependent mechanism promoting efficient, timed cyclin B degradation.
Collapse
Affiliation(s)
- Renaud Caous
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Aude Pascal
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Pierre Romé
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Laurent Richard-Parpaillon
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Roger Karess
- Institut Jacques Monod-Université Paris Diderot-Paris 7, 15 rue Hélène Brion, 75205 Paris, France
| | - Régis Giet
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| |
Collapse
|
163
|
Mutlu P, Ural AU, Gündüz U. Different types of cell cycle- and apoptosis-related gene expressions alter in corticosteroid-, vincristine-, and melphalan-resistant u-266 multiple myeloma cell lines. Turk J Haematol 2015; 31:231-8. [PMID: 25330516 PMCID: PMC4287023 DOI: 10.4274/tjh.2013.0231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Objective: Deregulation of the cell cycle and apoptosis mechanisms in normal cells causes many problems, including cancer. In this study, a genome-wide expression analysis of cell cycle- and apoptosis-related genes in corticosteroid-, vincristine-, and melphalan-resistant U-266 multiple myeloma cell lines was conducted. Materials and Methods: Resistant U-266 sublines were induced by application of each drug by stepwise dose increments. Resistance gained by the cells was confirmed with XTT cytotoxicity assay and microarray analyses were carried out. Among the cell cycle- and apoptosis-related gene expressions, alterations of more than 2-fold were considered significant. Results: Cyclin E2 was drastically overexpressed in the vincristine-resistant subline and a general upregulation was observed for various cyclin-dependent kinases. Some of the cyclin-dependent kinase inhibitor encoding genes were downregulated in resistant sublines in general. Tumor necrosis factor receptor genes were generally downregulated in corticosteroid- and melphalan-resistant U-266 sublines. Different types of effector caspases were downregulated in all resistant sublines. Ceramide metabolism genes seemed to be changed in favor of survival, especially in the melphalan-resistant subline. Conclusion: This report shows that different types of chemotherapeutic drugs alter different apoptotic and cell cycle-related gene expressions and, as a result, may cause drug-resistant phenotypes in U-266 multiple myeloma cell lines. Among those gene expressions, the most drastic increase in cyclin E2 could be important for the survival of vincristine-resistant U-266 cell lines, whereas alteration of ceramide metabolism genes could be important in melphalan resistance.
Collapse
Affiliation(s)
- Pelin Mutlu
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkey. E-ma-il:
| | | | | |
Collapse
|
164
|
Ramirez J, Martinez A, Lectez B, Lee SY, Franco M, Barrio R, Dittmar G, Mayor U. Proteomic Analysis of the Ubiquitin Landscape in the Drosophila Embryonic Nervous System and the Adult Photoreceptor Cells. PLoS One 2015; 10:e0139083. [PMID: 26460970 PMCID: PMC4604154 DOI: 10.1371/journal.pone.0139083] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/09/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Ubiquitination is known to regulate physiological neuronal functions as well as to be involved in a number of neuronal diseases. Several ubiquitin proteomic approaches have been developed during the last decade but, as they have been mostly applied to non-neuronal cell culture, very little is yet known about neuronal ubiquitination pathways in vivo. METHODOLOGY/PRINCIPAL FINDINGS Using an in vivo biotinylation strategy we have isolated and identified the ubiquitinated proteome in neurons both for the developing embryonic brain and for the adult eye of Drosophila melanogaster. Bioinformatic comparison of both datasets indicates a significant difference on the ubiquitin substrates, which logically correlates with the processes that are most active at each of the developmental stages. Detection within the isolated material of two ubiquitin E3 ligases, Parkin and Ube3a, indicates their ubiquitinating activity on the studied tissues. Further identification of the proteins that do accumulate upon interference with the proteasomal degradative pathway provides an indication of the proteins that are targeted for clearance in neurons. Last, we report the proof-of-principle validation of two lysine residues required for nSyb ubiquitination. CONCLUSIONS/SIGNIFICANCE These data cast light on the differential and common ubiquitination pathways between the embryonic and adult neurons, and hence will contribute to the understanding of the mechanisms by which neuronal function is regulated. The in vivo biotinylation methodology described here complements other approaches for ubiquitome study and offers unique advantages, and is poised to provide further insight into disease mechanisms related to the ubiquitin proteasome system.
Collapse
Affiliation(s)
- Juanma Ramirez
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
- Functional Genomics Unit, CIC bioGUNE, Derio, Spain
| | - Aitor Martinez
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
- Functional Genomics Unit, CIC bioGUNE, Derio, Spain
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Benoit Lectez
- Functional Genomics Unit, CIC bioGUNE, Derio, Spain
- Mollecular Cell Biology, Turku Centre for Biotechnology, Turku, Finland
| | - So Young Lee
- Functional Genomics Unit, CIC bioGUNE, Derio, Spain
| | - Maribel Franco
- Functional Genomics Unit, CIC bioGUNE, Derio, Spain
- Developmental Neurobiology, Institute of Neurosciences, CSIC/UMH, Sant Joan d’Alacant, Alicante, Spain
| | - Rosa Barrio
- Functional Genomics Unit, CIC bioGUNE, Derio, Spain
| | - Gunnar Dittmar
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
- Functional Genomics Unit, CIC bioGUNE, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain
- * E-mail:
| |
Collapse
|
165
|
Lin Z, Tan C, Qiu Q, Kong S, Yang H, Zhao F, Liu Z, Li J, Kong Q, Gao B, Barrett T, Yang GY, Zhang J, Fang D. Ubiquitin-specific protease 22 is a deubiquitinase of CCNB1. Cell Discov 2015; 1. [PMID: 27030811 PMCID: PMC4809424 DOI: 10.1038/celldisc.2015.28] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The elevated level of CCNB1 indicates more aggressive cancer and poor prognosis. However, the factors that cause CCNB1 upregulation remain enigmatic. Herein, we identify USP22 as a CCNB1 interactor and discover that both USP22 and CCNB1 are dramatically elevated with a strong positive correlation in colon cancer tissues. USP22 stabilizes CCNB1 by antagonizing proteasome-mediated degradation in a cell cycle-specific manner. Phosphorylation of USP22 by CDK1 enhances its activity in deubiquitinating CCNB1. The ubiquitin ligase anaphase-promoting complex (APC/C) targets USP22 for degradation by using the substrate adapter CDC20 during cell exit from M phase, presumably allowing CCNB1 degradation. Finally, we discover that USP22 knockdown leads to slower cell growth and reduced tumor size. Our study demonstrates that USP22 is a CCNB1 deubiquitinase, suggesting that targeting USP22 might be an effective approach to treat cancers with elevated CCNB1 expression.
Collapse
Affiliation(s)
- Zhenghong Lin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Can Tan
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Quan Qiu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sinyi Kong
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Heeyoung Yang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Fang Zhao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zhaojian Liu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jinping Li
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Qingfei Kong
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Beixue Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Terry Barrett
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guang-Yu Yang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jianing Zhang
- School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| |
Collapse
|
166
|
Min M, Mevissen TET, De Luca M, Komander D, Lindon C. Efficient APC/C substrate degradation in cells undergoing mitotic exit depends on K11 ubiquitin linkages. Mol Biol Cell 2015; 26:4325-32. [PMID: 26446837 PMCID: PMC4666129 DOI: 10.1091/mbc.e15-02-0102] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 09/29/2015] [Indexed: 11/11/2022] Open
Abstract
The ubiquitin proteasome system (UPS) directs programmed destruction of key cellular regulators via posttranslational modification of its targets with polyubiquitin chains. These commonly contain Lys-48 (K48)-directed ubiquitin linkages, but chains containing atypical Lys-11 (K11) linkages also target substrates to the proteasome--for example, to regulate cell cycle progression. The ubiquitin ligase called the anaphase-promoting complex/cyclosome (APC/C) controls mitotic exit. In higher eukaryotes, the APC/C works with the E2 enzyme UBE2S to assemble K11 linkages in cells released from mitotic arrest, and these are proposed to constitute an improved proteolytic signal during exit from mitosis. We tested this idea by correlating quantitative measures of in vivo K11-specific ubiquitination of individual substrates, including Aurora kinases, with their degradation kinetics tracked at the single-cell level. All anaphase substrates tested by this methodology are stabilized by depletion of K11 linkages via UBE2S knockdown, even if the same substrates are significantly modified with K48-linked polyubiquitin. Specific examination of substrates depending on the APC/C coactivator Cdh1 for their degradation revealed Cdh1-dependent enrichment of K11 chains on these substrates, whereas other ubiquitin linkages on the same substrates added during mitotic exit were Cdh1-independent. Therefore we show that K11 linkages provide the APC/C with a means to regulate the rate of substrate degradation in a coactivator-specified manner.
Collapse
Affiliation(s)
- Mingwei Min
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - Tycho E T Mevissen
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 OQH, United Kingdom
| | - Maria De Luca
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - David Komander
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 OQH, United Kingdom
| | - Catherine Lindon
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| |
Collapse
|
167
|
Defective sister chromatid cohesion is synthetically lethal with impaired APC/C function. Nat Commun 2015; 6:8399. [PMID: 26423134 PMCID: PMC4600715 DOI: 10.1038/ncomms9399] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 08/19/2015] [Indexed: 01/05/2023] Open
Abstract
Warsaw breakage syndrome (WABS) is caused by defective DDX11, a DNA helicase that is essential for chromatid cohesion. Here, a paired genome-wide siRNA screen in patient-derived cell lines reveals that WABS cells do not tolerate partial depletion of individual APC/C subunits or the spindle checkpoint inhibitor p31comet. A combination of reduced cohesion and impaired APC/C function also leads to fatal mitotic arrest in diploid RPE1 cells. Moreover, WABS cell lines, and several cancer cell lines with cohesion defects, display a highly increased response to a new cell-permeable APC/C inhibitor, apcin, but not to the spindle poison paclitaxel. Synthetic lethality of APC/C inhibition and cohesion defects strictly depends on a functional mitotic spindle checkpoint as well as on intact microtubule pulling forces. This indicates that the underlying mechanism involves cohesion fatigue in response to mitotic delay, leading to spindle checkpoint re-activation and lethal mitotic arrest. Our results point to APC/C inhibitors as promising therapeutic agents targeting cohesion-defective cancers. Cohesion is associated with many forms of cancer. De Lange et al. show that such cohesion defects can sensitise cells to apoptosis in response to a new APC/C ubiquitin ligase inhibitor, by prolonging mitotic arrest and checkpoint activation due to cohesion fatigue.
Collapse
|
168
|
Poli A, Ramazzotti G, Matteucci A, Manzoli L, Lonetti A, Suh PG, McCubrey JA, Cocco L. A novel DAG-dependent mechanism links PKCɑ and Cyclin B1 regulating cell cycle progression. Oncotarget 2015; 5:11526-40. [PMID: 25362646 PMCID: PMC4294327 DOI: 10.18632/oncotarget.2578] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/06/2014] [Indexed: 01/11/2023] Open
Abstract
Through the years, different studies showed the involvement of Protein Kinase C (PKC) in cell cycle control, in particular during G1/S transition. Little is known about their role at G2/M checkpoint. In this study, using K562 human erythroleukemia cell line, we found a novel and specific mechanism through which the conventional isoform PKC⍺ positively affects Cyclin B1 modulating G2/M progression of cell cycle. Since the kinase activity of this PKC isoform was not necessary in this process, we demonstrated that PKC⍺, physically interacting with Cyclin B1, avoided its degradation and stimulated its nuclear import at mitosis. Moreover, the process resulted to be strictly connected with the increase in nuclear diacylglycerol levels (DAG) at G2/M checkpoint, due to the activity of nuclear Phospholipase C β1 (PLCβ1), the only PLC isoform mainly localized in the nucleus of K562 cells. Taken together, our findings indicated a novel DAG dependent mechanism able to regulate the G2/M progression of the cell cycle.
Collapse
Affiliation(s)
- Alessandro Poli
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| | - Giulia Ramazzotti
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Matteucci
- CNR-National Research Council of Italy, Institute of Molecular Genetics, Bologna, Italy
| | - Lucia Manzoli
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| | - Annalisa Lonetti
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| | - Pann-Ghill Suh
- School of Nano-Biotechnology and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Lucio Cocco
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
169
|
Qian X, Song X, He Y, Yang Z, Sun T, Wang J, Zhu G, Xing W, You C. CCNB2 overexpression is a poor prognostic biomarker in Chinese NSCLC patients. Biomed Pharmacother 2015; 74:222-7. [PMID: 26349989 DOI: 10.1016/j.biopha.2015.08.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/30/2015] [Accepted: 08/02/2015] [Indexed: 12/27/2022] Open
Abstract
Cyclin B2 (CCNB2), a member of cyclin family proteins, serves a key role in progression of G2/M transition. The clinical value of CCNB2 in non-small cell lung cancer is still unknown. The aim of our study is to identify the role of CCNB2 in NSCLC patients. The status of CCNB2 in NSCLC tissues and normal lung tissues was observed in Gene Expression Omnibus (GEO: GSE19804). CCNB2 mRNA and protein expressions were detected in NSCLC and normal lung tissues by using Real-time PCR and immunohistochemistry. The association of CCNB protein expression with clinical characteristics of 186 NSCLC patients was analyzed by immunohistochemistry. Based on microarray data (GEO: GSE19804), we observed that CCNB2 was significantly overexpressed in NSCLC tissues compared with paired adjacent normal lung tissue. Furthermore, we verified mRNA and protein levels of CCNB2 expression were both increased in NSCLC tissues. We found high levels of CCNB2 protein were positively associated with the status of differentiated degree, tumor size, lymph node metastasis, distant metastasis, and clinical stage. Meanwhile, CCNB2 protein overexpression was an independent unfavorable prognostic factor for the overall survival of patients with NSCLC. In conclusion, overexpression of CCNB2 protein is associated with clinical progression and poor prognosis in NSCLC.
Collapse
Affiliation(s)
- Xiaotao Qian
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xuekun Song
- School of Information Technology, Henan University of Traditional Chinese Medicine, Zhengzhou450008, China
| | - Yuan He
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhiyong Yang
- Department of Radiontherapy, Huanggang Central Hospital, Huanggang 438000, China
| | - Tao Sun
- Department of Thoracic Surgery, Fuyang Second People's Hospital, Fuyang 236015, China
| | - Jing Wang
- Department of Oncology, Fuyang Tumor Hospital, Fuyang 236018, China
| | - Guiqi Zhu
- Department of Infection and Liver Diseases, Liver Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Weihai Xing
- Department of Pathology, Fifth People's Hospital of Fuyang, Fuyang 23600, China
| | - Changxuan You
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
170
|
Tracking the Cyclin B1-GFP Sensor to Profile the Pattern of Mitosis Versus Mitotic Bypass. Methods Mol Biol 2015; 1342:279-85. [PMID: 26254931 DOI: 10.1007/978-1-4939-2957-3_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
This chapter provides a method for quantitative single cell analysis to track the transition of single cells from G2, indicated by high cyclin B1 levels, to G1 polyploidy phase (G1(p)), indicated by low cyclin B1 levels, in a 4n population. The cell tracking methodology described provides a fluorescence fingerprint suitable for deriving G2/M or G2/G1p transitions. Notably, during late G2 the absolute cyclin B1-eGFP reporter levels obtained were high and the switch-off point identifiable, with destruction rates of a similar order across all cell cycle routing avenues. The three principle parameters extracted were defined as (1) G2-to-G1(p) transition duration (tGFP(off)); (2) rate of sensor destruction (kGFP(off)), and (3) peak sensor expression (GFP(peak)).
Collapse
|
171
|
Nguyen LK. Dynamics of ubiquitin-mediated signalling: insights from mathematical modelling and experimental studies. Brief Bioinform 2015. [DOI: 10.1093/bib/bbv052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
172
|
Delacour Q, Li C, Plamont MA, Billon-Denis E, Aujard I, Le Saux T, Jullien L, Gautier A. Light-Activated Proteolysis for the Spatiotemporal Control of Proteins. ACS Chem Biol 2015; 10:1643-7. [PMID: 25938742 DOI: 10.1021/acschembio.5b00069] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The regulation of proteolysis is an efficient way to control protein function in cells. Here, we present a general strategy enabling to increase the spatiotemporal resolution of conditional proteolysis by using light activation as trigger. Our approach relies on the auxin-inducible degradation system obtained by transposing components of the plant auxin-dependent degradation pathway in mammalian cells. We developed a photoactivatable auxin that acts as a photoactivatable inducer of degradation. Upon local and short light illumination, auxin is released in cells and triggers the degradation of a protein of interest with spatiotemporal control.
Collapse
Affiliation(s)
- Quentin Delacour
- Department
of Chemistry, École Normale Supérieure−PSL Research University, 24 rue Lhomond, F-75005 Paris, France
- Sorbonne Universités, UPMC
Univ Paris 06, UMR 8640 PASTEUR, F-75005 Paris, France
- CNRS, UMR 8640 PASTEUR, F-75005 Paris, France
| | - Chenge Li
- Department
of Chemistry, École Normale Supérieure−PSL Research University, 24 rue Lhomond, F-75005 Paris, France
- Sorbonne Universités, UPMC
Univ Paris 06, UMR 8640 PASTEUR, F-75005 Paris, France
- CNRS, UMR 8640 PASTEUR, F-75005 Paris, France
| | - Marie-Aude Plamont
- Department
of Chemistry, École Normale Supérieure−PSL Research University, 24 rue Lhomond, F-75005 Paris, France
- Sorbonne Universités, UPMC
Univ Paris 06, UMR 8640 PASTEUR, F-75005 Paris, France
- CNRS, UMR 8640 PASTEUR, F-75005 Paris, France
| | - Emmanuelle Billon-Denis
- Department
of Chemistry, École Normale Supérieure−PSL Research University, 24 rue Lhomond, F-75005 Paris, France
- Sorbonne Universités, UPMC
Univ Paris 06, UMR 8640 PASTEUR, F-75005 Paris, France
- CNRS, UMR 8640 PASTEUR, F-75005 Paris, France
| | - Isabelle Aujard
- Department
of Chemistry, École Normale Supérieure−PSL Research University, 24 rue Lhomond, F-75005 Paris, France
- Sorbonne Universités, UPMC
Univ Paris 06, UMR 8640 PASTEUR, F-75005 Paris, France
- CNRS, UMR 8640 PASTEUR, F-75005 Paris, France
| | - Thomas Le Saux
- Department
of Chemistry, École Normale Supérieure−PSL Research University, 24 rue Lhomond, F-75005 Paris, France
- Sorbonne Universités, UPMC
Univ Paris 06, UMR 8640 PASTEUR, F-75005 Paris, France
- CNRS, UMR 8640 PASTEUR, F-75005 Paris, France
| | - Ludovic Jullien
- Department
of Chemistry, École Normale Supérieure−PSL Research University, 24 rue Lhomond, F-75005 Paris, France
- Sorbonne Universités, UPMC
Univ Paris 06, UMR 8640 PASTEUR, F-75005 Paris, France
- CNRS, UMR 8640 PASTEUR, F-75005 Paris, France
| | - Arnaud Gautier
- Department
of Chemistry, École Normale Supérieure−PSL Research University, 24 rue Lhomond, F-75005 Paris, France
- Sorbonne Universités, UPMC
Univ Paris 06, UMR 8640 PASTEUR, F-75005 Paris, France
- CNRS, UMR 8640 PASTEUR, F-75005 Paris, France
| |
Collapse
|
173
|
Milev MP, Hasaj B, Saint-Dic D, Snounou S, Zhao Q, Sacher M. TRAMM/TrappC12 plays a role in chromosome congression, kinetochore stability, and CENP-E recruitment. ACTA ACUST UNITED AC 2015; 209:221-34. [PMID: 25918224 PMCID: PMC4411272 DOI: 10.1083/jcb.201501090] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The TRAPP subunit TrappC12/TTC15, here renamed TRAMM, plays a role in the regulation of kinetochore stability and CENP-E recruitment during mitosis. Chromosome congression requires the stable attachment of microtubules to chromosomes mediated by the kinetochore, a large proteinaceous structure whose mechanism of assembly is unknown. In this paper, we present the finding that a protein called TRAMM (formerly known as TrappC12) plays a role in mitosis. Depletion of TRAMM resulted in noncongressed chromosomes and arrested cells in mitosis. Small amounts of TRAMM associated with chromosomes, and its depletion affected the localization of some kinetochore proteins, the strongest effect being seen for CENP-E. TRAMM interacts with CENP-E, and depletion of TRAMM prevented the recruitment of CENP-E to the kinetochore. TRAMM is phosphorylated early in mitosis and dephosphorylated at the onset of anaphase. Interestingly, this phosphorylation/dephosphorylation cycle correlates with its association/disassociation with CENP-E. Finally, we demonstrate that a phosphomimetic form of TRAMM recruited CENP-E to kinetochores more efficiently than did the nonphosphorylatable mutant. Our study identifies a moonlighting function for TRAMM during mitosis and adds a new component that regulates kinetochore stability and CENP-E recruitment.
Collapse
Affiliation(s)
- Miroslav P Milev
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Benedeta Hasaj
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Djenann Saint-Dic
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Sary Snounou
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Qingchuan Zhao
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Michael Sacher
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
| |
Collapse
|
174
|
Abstract
A close relationship between proliferation and cell fate specification has been well documented in many developmental systems. In addition to the gradual cell fate changes accompanying normal development and tissue homeostasis, it is now commonly appreciated that cell fate could also undergo drastic changes, as illustrated by the induction of pluripotency from many differentiated somatic cell types during the process of Yamanaka reprogramming. Strikingly, the drastic cell fate change induced by Yamanaka factors (Oct4, Sox2, Klf4, and c-Myc) is preceded by extensive cell cycle acceleration. Prompted by our recent discovery that progression toward pluripotency from rare somatic cells could bypass the stochastic phase of reprogramming and that a key feature of these somatic cells is an ultrafast cell cycle (~8 h/cycle), we assess whether cell cycle dynamics could provide a general framework for controlling cell fate. Several potential mechanisms on how cell cycle dynamics may impact cell fate determination by regulating chromatin, key transcription factor concentration, or their interactions are discussed. Specific challenges and implications for studying and manipulating cell fate are considered.
Collapse
|
175
|
Whalley HJ, Porter AP, Diamantopoulou Z, White GRM, Castañeda-Saucedo E, Malliri A. Cdk1 phosphorylates the Rac activator Tiam1 to activate centrosomal Pak and promote mitotic spindle formation. Nat Commun 2015; 6:7437. [PMID: 26078008 PMCID: PMC4490568 DOI: 10.1038/ncomms8437] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 05/08/2015] [Indexed: 01/13/2023] Open
Abstract
Centrosome separation is critical for bipolar spindle formation and the accurate segregation of chromosomes during mammalian cell mitosis. Kinesin-5 (Eg5) is a microtubule motor essential for centrosome separation, and Tiam1 and its substrate Rac antagonize Eg5-dependent centrosome separation in early mitosis promoting efficient chromosome congression. Here we identify S1466 of Tiam1 as a novel Cdk1 site whose phosphorylation is required for the mitotic function of Tiam1. We find that this phosphorylation of Tiam1 is required for the activation of group I p21-activated kinases (Paks) on centrosomes in prophase. Further, we show that both Pak1 and Pak2 counteract centrosome separation in a kinase-dependent manner and demonstrate that they act downstream of Tiam1. We also show that depletion of Pak1/2 allows cells to escape monopolar arrest by Eg5 inhibition, highlighting the potential importance of this signalling pathway for the development of Eg5 inhibitors as cancer therapeutics.
Collapse
Affiliation(s)
- Helen J. Whalley
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Andrew P. Porter
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Zoi Diamantopoulou
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Gavin R. M. White
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Eduardo Castañeda-Saucedo
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Angeliki Malliri
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| |
Collapse
|
176
|
Gera N, Yang A, Holtzman TS, Lee SX, Wong ET, Swanson KD. Tumor treating fields perturb the localization of septins and cause aberrant mitotic exit. PLoS One 2015; 10:e0125269. [PMID: 26010837 PMCID: PMC4444126 DOI: 10.1371/journal.pone.0125269] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 03/23/2015] [Indexed: 01/04/2023] Open
Abstract
The anti-tumor effects of chemotherapy and radiation are thought to be mediated by triggering G1/S or G2/M cell cycle checkpoints, while spindle poisons, such as paclitaxel, block metaphase exit by initiating the spindle assembly checkpoint. In contrast, we have found that 150 kilohertz (kHz) alternating electric fields, also known as Tumor Treating Fields (TTFields), perturbed cells at the transition from metaphase to anaphase. Cells exposed to the TTFields during mitosis showed normal progression to this point, but exhibited uncontrolled membrane blebbing that coincided with metaphase exit. The ability of such alternating electric fields to affect cellular physiology is likely to be dependent on their interactions with proteins possessing high dipole moments. The mitotic Septin complex consisting of Septin 2, 6 and 7, possesses a high calculated dipole moment of 2711 Debyes (D) and plays a central role in positioning the cytokinetic cleavage furrow, and governing its contraction during ingression. We showed that during anaphase, TTFields inhibited Septin localization to the anaphase spindle midline and cytokinetic furrow, as well as its association with microtubules during cell attachment and spreading on fibronectin. After aberrant metaphase exit as a consequence of TTFields exposure, cells exhibited aberrant nuclear architecture and signs of cellular stress including an overall decrease in cellular proliferation, followed by apoptosis that was strongly influenced by the p53 mutational status. Thus, TTFields are able to diminish cell proliferation by specifically perturbing key proteins involved in cell division, leading to mitotic catastrophe and subsequent cell death.
Collapse
Affiliation(s)
- Nidhi Gera
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aaron Yang
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Talia S. Holtzman
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Sze Xian Lee
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Eric T. Wong
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Kenneth D. Swanson
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
177
|
CDK1 structures reveal conserved and unique features of the essential cell cycle CDK. Nat Commun 2015; 6:6769. [PMID: 25864384 PMCID: PMC4413027 DOI: 10.1038/ncomms7769] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/26/2015] [Indexed: 12/23/2022] Open
Abstract
CDK1 is the only essential cell cycle CDK in human cells and is required for successful completion of M-phase. It is the founding member of the CDK family and is conserved across all eukaryotes. Here we report the crystal structures of complexes of CDK1–Cks1 and CDK1–cyclin B–Cks2. These structures confirm the conserved nature of the inactive monomeric CDK fold and its ability to be remodelled by cyclin binding. Relative to CDK2–cyclin A, CDK1–cyclin B is less thermally stable, has a smaller interfacial surface, is more susceptible to activation segment dephosphorylation and shows differences in the substrate sequence features that determine activity. Both CDK1 and CDK2 are potential cancer targets for which selective compounds are required. We also describe the first structure of CDK1 bound to a potent ATP-competitive inhibitor and identify aspects of CDK1 structure and plasticity that might be exploited to develop CDK1-selective inhibitors. Cyclin-dependent kinases are the principal drivers of cell cycle progression. Here the authors present several crystal structures of Cdk1 in complex with cyclin B and/or the assembly factors Cks1/2 and a small molecule inhibitor to reveal key features of this essential mitotic kinase.
Collapse
|
178
|
Targeting Cdc20 as a novel cancer therapeutic strategy. Pharmacol Ther 2015; 151:141-51. [PMID: 25850036 DOI: 10.1016/j.pharmthera.2015.04.002] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 03/31/2015] [Indexed: 12/25/2022]
Abstract
The Anaphase Promoting Complex (APC, also called APC/C) regulates cell cycle progression by forming two closely related, but functionally distinct E3 ubiquitin ligase sub-complexes, APC(Cdc20) and APC(Cdh1), respectively. Emerging evidence has begun to reveal that Cdc20 and Cdh1 have opposing functions in tumorigenesis. Specifically, Cdh1 functions largely as a tumor suppressor, whereas Cdc20 exhibits an oncogenic function, suggesting that Cdc20 could be a promising therapeutic target for combating human cancer. However, the exact underlying molecular mechanisms accounting for their differences in tumorigenesis remain largely unknown. Therefore, in this review, we summarize the downstream substrates of Cdc20 and the critical functions of Cdc20 in cell cycle progression, apoptosis, ciliary disassembly and brain development. Moreover, we briefly describe the upstream regulators of Cdc20 and the oncogenic role of Cdc20 in a variety of human malignancies. Furthermore, we summarize multiple pharmacological Cdc20 inhibitors including TAME and Apcin, and their potential clinical benefits. Taken together, development of specific Cdc20 inhibitors could be a novel strategy for the treatment of human cancers with elevated Cdc20 expression.
Collapse
|
179
|
Ishihara K, Nguyen PA, Wühr M, Groen AC, Field CM, Mitchison TJ. Organization of early frog embryos by chemical waves emanating from centrosomes. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0454. [PMID: 25047608 DOI: 10.1098/rstb.2013.0454] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The large cells in early vertebrate development face an extreme physical challenge in organizing their cytoplasm. For example, amphibian embryos have to divide cytoplasm that spans hundreds of micrometres every 30 min according to a precise geometry, a remarkable accomplishment given the extreme difference between molecular and cellular scales in this system. How do the biochemical reactions occurring at the molecular scale lead to this emergent behaviour of the cell as a whole? Based on recent findings, we propose that the centrosome plays a crucial role by initiating two autocatalytic reactions that travel across the large cytoplasm as chemical waves. Waves of mitotic entry and exit propagate out from centrosomes using the Cdk1 oscillator to coordinate the timing of cell division. Waves of microtubule-stimulated microtubule nucleation propagate out to assemble large asters that position spindles for the following mitosis and establish cleavage plane geometry. By initiating these chemical waves, the centrosome rapidly organizes the large cytoplasm during the short embryonic cell cycle, which would be impossible using more conventional mechanisms such as diffusion or nucleation by structural templating. Large embryo cells provide valuable insights to how cells control chemical waves, which may be a general principle for cytoplasmic organization.
Collapse
Affiliation(s)
- Keisuke Ishihara
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Phuong A Nguyen
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Martin Wühr
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Aaron C Groen
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Christine M Field
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Timothy J Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| |
Collapse
|
180
|
Sivakumar S, Gorbsky GJ. Spatiotemporal regulation of the anaphase-promoting complex in mitosis. Nat Rev Mol Cell Biol 2015; 16:82-94. [PMID: 25604195 DOI: 10.1038/nrm3934] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The appropriate timing of events that lead to chromosome segregation during mitosis and cytokinesis is essential to prevent aneuploidy, and defects in these processes can contribute to tumorigenesis. Key mitotic regulators are controlled through ubiquitylation and proteasome-mediated degradation. The APC/C (anaphase-promoting complex; also known as the cyclosome) is an E3 ubiquitin ligase that has a crucial function in the regulation of the mitotic cell cycle, particularly at the onset of anaphase and during mitotic exit. Co-activator proteins, inhibitor proteins, protein kinases and phosphatases interact with the APC/C to temporally and spatially control its activity and thus ensure accurate timing of mitotic events.
Collapse
Affiliation(s)
- Sushama Sivakumar
- Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, Oklahoma 73104, USA
| | - Gary J Gorbsky
- Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
181
|
Gerhold AR, Ryan J, Vallée-Trudeau JN, Dorn JF, Labbé JC, Maddox PS. Investigating the regulation of stem and progenitor cell mitotic progression by in situ imaging. Curr Biol 2015; 25:1123-34. [PMID: 25819563 DOI: 10.1016/j.cub.2015.02.054] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/15/2015] [Accepted: 02/19/2015] [Indexed: 10/23/2022]
Abstract
Genome stability relies upon efficacious chromosome congression and regulation by the spindle assembly checkpoint (SAC). The study of these fundamental mitotic processes in adult stem and progenitor cells has been limited by the technical challenge of imaging mitosis in these cells in situ. Notably, how broader physiological changes, such as dietary intake or age, affect mitotic progression in stem and/or progenitor cells is largely unknown. Using in situ imaging of C. elegans adult germlines, we describe the mitotic parameters of an adult stem and progenitor cell population in an intact animal. We find that SAC regulation in germline stem and progenitor cells is distinct from that found in early embryonic divisions and is more similar to that of classical tissue culture models. We further show that changes in organismal physiology affect mitotic progression in germline stem and progenitor cells. Reducing dietary intake produces a checkpoint-dependent delay in anaphase onset, and inducing dietary restriction when the checkpoint is impaired increases the incidence of segregation errors in mitotic and meiotic cells. Similarly, developmental aging of the germline stem and progenitor cell population correlates with a decline in the rate of several mitotic processes. These results provide the first in vivo validation of models for SAC regulation developed in tissue culture systems and demonstrate that several fundamental features of mitotic progression in adult stem and progenitor cells are highly sensitive to organismal physiological changes.
Collapse
Affiliation(s)
- Abigail R Gerhold
- Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Joël Ryan
- Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Julie-Nathalie Vallée-Trudeau
- Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Jonas F Dorn
- Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Jean-Claude Labbé
- Institute of Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada; Department of Pathology and Cell Biology, Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada.
| | - Paul S Maddox
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
182
|
Wang H, Li R, Li L, Ge Z, Zhou R, Li R. LRD-22, a novel dual dithiocarbamatic acid ester, inhibits Aurora-A kinase and induces apoptosis and cell cycle arrest in HepG2 cells. Biochem Biophys Res Commun 2015; 458:201-7. [PMID: 25645017 DOI: 10.1016/j.bbrc.2015.01.102] [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: 01/16/2015] [Accepted: 01/21/2015] [Indexed: 10/24/2022]
Abstract
In this study we investigated the antitumor activity of the novel dual dithiocarbamatic acid ester LRD-22 in vitro and in vivo. Several cancer cell lines were employed to determine the effect of LRD-22 on cell growth, and the MTT assay showed there was a significant decrease in viable tumor cell numbers in the presence of LRD-22, especially in the HepG2 cell line. Colony formation assay also showed LRD-22 strongly inhibits HepG2 cell growth. Evaluation of the mechanism involved showed that inhibitory effects of LRD-22 on cell growth are due to induction of apoptosis and G2/M arrest. LRD-22 inhibited Aurora-A phosphorylation at Thr288 and subsequently impaired p53 phosphorylation at Ser315 which was associated with the proteasome degradation pathway. Tumor suppressor protein p53 is stabilized by this mechanism and accumulates through inhibition of Aurora-A kinase activity via treatment with LRD-22. In vivo study of HepG2 xenograft in nude mice also shows LRD-22 suppresses tumor growth at a concentration of 5 mg/kg without animals suffering loss of body weight. In conclusion, our results demonstrate LRD-22 acts as an Aurora-A kinase inhibitor to induce apoptosis and inhibit proliferation in HepG2 cells, and should be considered as a promising targeting agent for HCC therapy.
Collapse
Affiliation(s)
- Huiling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, PR China
| | - Ridong Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, PR China
| | - Li Li
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, PR China
| | - Zemei Ge
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, PR China
| | - Rouli Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, PR China.
| | - Runtao Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, PR China.
| |
Collapse
|
183
|
Di Fiore B, Davey NE, Hagting A, Izawa D, Mansfeld J, Gibson TJ, Pines J. The ABBA motif binds APC/C activators and is shared by APC/C substrates and regulators. Dev Cell 2015; 32:358-372. [PMID: 25669885 PMCID: PMC4713905 DOI: 10.1016/j.devcel.2015.01.003] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 09/03/2014] [Accepted: 01/05/2015] [Indexed: 11/30/2022]
Abstract
The anaphase-promoting complex or cyclosome (APC/C) is the ubiquitin ligase that regulates mitosis by targeting specific proteins for degradation at specific times under the control of the spindle assembly checkpoint (SAC). How the APC/C recognizes its different substrates is a key problem in the control of cell division. Here, we have identified the ABBA motif in cyclin A, BUBR1, BUB1, and Acm1, and we show that it binds to the APC/C coactivator CDC20. The ABBA motif in cyclin A is required for its proper degradation in prometaphase through competing with BUBR1 for the same site on CDC20. Moreover, the ABBA motifs in BUBR1 and BUB1 are necessary for the SAC to work at full strength and to recruit CDC20 to kinetochores. Thus, we have identified a conserved motif integral to the proper control of mitosis that connects APC/C substrate recognition with the SAC.
Collapse
Affiliation(s)
- Barbara Di Fiore
- The Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Norman E. Davey
- Department of Physiology and Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Baden-Württemberg 69117, Germany
| | - Anja Hagting
- The Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Daisuke Izawa
- The Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Jörg Mansfeld
- Technische Universität Dresden, Tatzberg 47/49, 01307 Dresden, Germany
| | - Toby J. Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Baden-Württemberg 69117, Germany
| | - Jonathon Pines
- The Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge, CB2 1QN, UK
| |
Collapse
|
184
|
Abstract
In this article, we will discuss the biochemistry of mitosis in eukaryotic cells. We will focus on conserved principles that, importantly, are adapted to the biology of the organism. It is vital to bear in mind that the structural requirements for division in a rapidly dividing syncytial Drosophila embryo, for example, are markedly different from those in a unicellular yeast cell. Nevertheless, division in both systems is driven by conserved modules of antagonistic protein kinases and phosphatases, underpinned by ubiquitin-mediated proteolysis, which create molecular switches to drive each stage of division forward. These conserved control modules combine with the self-organizing properties of the subcellular architecture to meet the specific needs of the cell. Our discussion will draw on discoveries in several model systems that have been important in the long history of research on mitosis, and we will try to point out those principles that appear to apply to all cells, compared with those in which the biochemistry has been specifically adapted in a particular organism.
Collapse
Affiliation(s)
- Samuel Wieser
- The Gurdon Institute, Cambridge CB2 1QN, United Kingdom
| | | |
Collapse
|
185
|
Roukos V, Pegoraro G, Voss TC, Misteli T. Cell cycle staging of individual cells by fluorescence microscopy. Nat Protoc 2015; 10:334-48. [PMID: 25633629 PMCID: PMC6318798 DOI: 10.1038/nprot.2015.016] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Progression through the cell cycle is one of the most fundamental features of cells. Studies of the cell cycle have traditionally relied on the analysis of populations, and they often require specific markers or the use of genetically modified systems, making it difficult to determine the cell cycle stage of individual, unperturbed cells. We describe a protocol, suitable for use in high-resolution imaging approaches, for determining cell cycle staging of individual cells by measuring their DNA content by fluorescence microscopy. The approach is based on the accurate quantification by image analysis of the integrated nuclear intensity of cells stained with a DNA dye, and it can be used in combination with several histochemical methods. We describe and provide the algorithms for two automated image analysis pipelines and the derivation of cell cycle profiles with both commercial and open-source software. This 1-2-d protocol is applicable to adherent cells, and it is adaptable for use with several DNA dyes.
Collapse
Affiliation(s)
- Vassilis Roukos
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Gianluca Pegoraro
- 1] National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA. [2] High-Throughput Imaging Facility, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Ty C Voss
- 1] National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA. [2] PerkinElmer Health Sciences Inc., Waltham, Maryland, USA
| | - Tom Misteli
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
186
|
Izawa D, Pines J. The mitotic checkpoint complex binds a second CDC20 to inhibit active APC/C. Nature 2015; 517:631-4. [PMID: 25383541 PMCID: PMC4312099 DOI: 10.1038/nature13911] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 10/02/2014] [Indexed: 12/23/2022]
Abstract
The spindle assembly checkpoint (SAC) maintains genomic stability by delaying chromosome segregation until the last chromosome has attached to the mitotic spindle. The SAC prevents the anaphase promoting complex/cyclosome (APC/C) ubiquitin ligase from recognizing cyclin B and securin by catalysing the incorporation of the APC/C co-activator, CDC20, into a complex called the mitotic checkpoint complex (MCC). The SAC works through unattached kinetochores generating a diffusible 'wait anaphase' signal that inhibits the APC/C in the cytoplasm, but the nature of this signal remains a key unsolved problem. Moreover, the SAC and the APC/C are highly responsive to each other: the APC/C quickly targets cyclin B and securin once all the chromosomes attach in metaphase, but is rapidly inhibited should kinetochore attachment be perturbed. How this is achieved is also unknown. Here, we show that the MCC can inhibit a second CDC20 that has already bound and activated the APC/C. We show how the MCC inhibits active APC/C and that this is essential for the SAC. Moreover, this mechanism can prevent anaphase in the absence of kinetochore signalling. Thus, we propose that the diffusible 'wait anaphase' signal could be the MCC itself, and explain how reactivating the SAC can rapidly inhibit active APC/C.
Collapse
Affiliation(s)
- Daisuke Izawa
- The Gurdon Institute and Department of Zoology, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Jonathon Pines
- The Gurdon Institute and Department of Zoology, Tennis Court Road, Cambridge CB2 1QN, UK
| |
Collapse
|
187
|
Tamura N, Simon JE, Nayak A, Shenoy RT, Hiroi N, Boilot V, Funahashi A, Draviam VM. A proteomic study of mitotic phase-specific interactors of EB1 reveals a role for SXIP-mediated protein interactions in anaphase onset. Biol Open 2015; 4:155-69. [PMID: 25596275 PMCID: PMC4365484 DOI: 10.1242/bio.201410413] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/07/2014] [Indexed: 12/12/2022] Open
Abstract
Microtubules execute diverse mitotic events that are spatially and temporally separated; the underlying regulation is poorly understood. By combining drug treatments, large-scale immunoprecipitation and mass spectrometry, we report the first comprehensive map of mitotic phase-specific protein interactions of the microtubule-end binding protein, EB1. EB1 interacts with some, but not all, of its partners throughout mitosis. We show that the interaction of EB1 with Astrin-SKAP complex, a key regulator of chromosome segregation, is enhanced during prometaphase, compared to anaphase. We find that EB1 and EB3, another EB family member, can interact directly with SKAP, in an SXIP-motif dependent manner. Using an SXIP defective mutant that cannot interact with EB, we uncover two distinct pools of SKAP at spindle microtubules and kinetochores. We demonstrate the importance of SKAP's SXIP-motif in controlling microtubule growth rates and anaphase onset, without grossly disrupting spindle function. Thus, we provide the first comprehensive map of temporal changes in EB1 interactors during mitosis and highlight the importance of EB protein interactions in ensuring normal mitosis.
Collapse
Affiliation(s)
- Naoka Tamura
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Judith E Simon
- Department of Genetics, University of Cambridge, Cambridge, UK Present address: European Research Institute for the Biology of Ageing, University of Groningen, Groningen, Netherlands
| | - Arnab Nayak
- Department of Genetics, University of Cambridge, Cambridge, UK Present address: Institute for Biochemistry II, Goethe University Frankfurt am Main, Germany
| | - Rajesh T Shenoy
- Department of Genetics, University of Cambridge, Cambridge, UK
| | | | - Viviane Boilot
- Department of Genetics, University of Cambridge, Cambridge, UK
| | | | - Viji M Draviam
- Department of Genetics, University of Cambridge, Cambridge, UK
| |
Collapse
|
188
|
Boekhout M, Wolthuis R. Nek2A destruction marks APC/C activation at the prophase-to-prometaphase transition by spindle-checkpoint restricted Cdc20. J Cell Sci 2015; 128:1639-53. [DOI: 10.1242/jcs.163279] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 02/06/2015] [Indexed: 12/31/2022] Open
Abstract
Nek2A is a presumed APC/CCdc20 substrate, which, like cyclin A, is degraded in mitosis while the spindle checkpoint is active. Cyclin A prevents spindle checkpoint proteins from binding to Cdc20 and is recruited to the APC/C in prometaphase. We found that Nek2A and cyclin A avoid stabilization by the spindle checkpoint in different ways. First, enhancing mitotic checkpoint complex (MCC) formation by nocodazole treatment inhibited the degradation of geminin and cyclin A while Nek2A disappeared at normal rate. Secondly, depleting Cdc20 effectively stabilized cyclin A but not Nek2A. Nevertheless, Nek2A destruction critically depended on Cdc20 binding to the APC/C. Thirdly, in contrast to cyclin A, Nek2A was recruited to the APC/C before the start of mitosis. Interestingly, the spindle checkpoint very effectively stabilized an APC/C-binding mutant of Nek2A, which required the Nek2A KEN box. Apparently, in cells, the spindle checkpoint primarily prevents Cdc20 from binding destruction motifs. Nek2A disappearance marks the prophase-to-prometaphase transition, when Cdc20, regardless of the spindle checkpoint, activates the APC/C. However, Mad2 depletion accelerated Nek2A destruction, showing that spindle checkpoint release further increases APC/CCdc20 catalytic activity.
Collapse
|
189
|
Raab M, Krämer A, Hehlgans S, Sanhaji M, Kurunci-Csacsko E, Dötsch C, Bug G, Ottmann O, Becker S, Pachl F, Kuster B, Strebhardt K. Mitotic arrest and slippage induced by pharmacological inhibition of Polo-like kinase 1. Mol Oncol 2015; 9:140-54. [PMID: 25169932 PMCID: PMC5528686 DOI: 10.1016/j.molonc.2014.07.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/25/2014] [Indexed: 01/22/2023] Open
Abstract
Exposure to drugs that interfere with microtubule dynamics block cell cycle progression at mitosis by prolonged activation of the spindle assembly checkpoint (SAC). Cells can evade mitotic arrest and proceed to interphase without chromosome segregation by a process termed mitotic slippage that involves Cyclin B1 degradation without checkpoint inactivation. Here, we explored the cellular response to small-molecule inhibitors of Polo-like kinase 1 (Plk1), an important regulator of cell division. We found that the clinical Plk1 inhibitors BI 2536 and BI 6727, both unexpectedly, induced a dose-dependent cellular drug response: While mitotic arrest was induced in cancer cell lines and primary non-transformed cells across the entire range of concentrations tested, only high concentrations seemed to promote mitotic slippage. Since this observation contrasts with the effects expected from studies reporting RNAi-mediated Plk1 depletion in cancer cells, we wondered whether both ATP-competitive inhibitors target unknown kinases that are involved in signaling from the spindle assembly checkpoint (SAC) and might contribute to the mitotic slippage. A chemical proteomics approach used to profile the selectivity of both inhibitors revealed that SAC kinases are not targeted directly. Still, the activities of Cdk1/Cyclin B1 and Aurora B, which plays important roles in the error correction of false microtubule-kinetochore attachments and in checkpoint signaling, were shown to be downregulated at high inhibitor concentrations. Our data suggest that the inhibition of Plk1 activity below a certain threshold influences Aurora B activity via reduced phosphorylation of Fox M1 and Survivin leading to diminished levels of Aurora B protein and alteration of its subcellular localization. Within the spectrum of SAC proteins that are degraded during mitotic slippage, the degradation of Cyclin B1 and the downregulation of Aurora B activity by Plk1 inhibition seem to be critical promoters of mitotic slippage. The results indicate that careful dose-finding studies in cancer trials are necessary to limit or even prevent mitotic slippage, which could be associated with improved cancer cell survival.
Collapse
Affiliation(s)
- Monika Raab
- Department of Gynecology, School of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Andrea Krämer
- Department of Gynecology, School of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | | - Mourad Sanhaji
- Department of Gynecology, School of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Elisabeth Kurunci-Csacsko
- Department of Gynecology, School of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Christina Dötsch
- Department of Gynecology, School of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Gesine Bug
- Department of Medicine, Hematology/Oncology, Goethe University, Germany
| | - Oliver Ottmann
- Department of Medicine, Hematology/Oncology, Goethe University, Germany
| | - Sven Becker
- Department of Gynecology, School of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Fiona Pachl
- Technische Universität München, Emil Erlenmeyer Forum 5, 85354 Freising, Germany
| | - Bernhard Kuster
- Technische Universität München, Emil Erlenmeyer Forum 5, 85354 Freising, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Klaus Strebhardt
- Department of Gynecology, School of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
190
|
El Gaafary M, Büchele B, Syrovets T, Agnolet S, Schneider B, Schmidt CQ, Simmet T. An α-acetoxy-tirucallic acid isomer inhibits Akt/mTOR signaling and induces oxidative stress in prostate cancer cells. J Pharmacol Exp Ther 2015; 352:33-42. [PMID: 25316122 DOI: 10.1124/jpet.114.217323] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Here we provide evidence that αATA(8,24) (3α-acetyloxy-tir-8,24-dien-21-oic acid) inhibits Akt/mammalian target of rapamycin (mTOR) signaling. αATA(8,24) and other tirucallic acids were isolated from the acetylated extract of the oleo gum resin of Boswellia serrata to chemical homogeneity. Compared with related tirucallic acids, αATA(8,24) was the most potent inhibitor of the proliferation of androgen-insensitive prostate cancer cells in vitro and in vivo, in prostate cancer xenografted onto chick chorioallantoic membranes. αATA(8,24) induced loss of cell membrane asymmetry, caspase-3 activation, and DNA fragmentation in vitro and in vivo. These effects were selective for cancer cells, because αATA(8,24) exerted no overt toxic effects on peripheral blood mononuclear cells or the chick embryo. At the molecular level, αATA(8,24) inhibited the Akt1 kinase activity. Prior to all biochemical signs of cellular dysfunction, αATA(8,24) induced inhibition of the Akt downstream target mTOR as indicated by dephosphorylation of S6K1. This event was followed by decreased expression of cell cycle regulators, such as cyclin D1, cyclin E, and cyclin B1, as well as cyclin-dependent kinases CDK4 and CDK2 and phosphoretinoblastoma protein, which led to inhibition of the cell-cycle progression. In agreement with the mTOR inhibition, αATA(8,24) and rapamycin increased the volume of acidic vesicular organelles. In contrast to rapamycin, αATA(8,24) destabilized lysosomal and mitochondrial membranes and induced reactive oxygen species production in cancer cells. The ability of αATA(8,24) to inhibit Akt/mTOR signaling and to induce simultaneously oxidative stress could be exploited for the development of novel antitumor therapeutics with a lower profile of toxic side effects.
Collapse
Affiliation(s)
- Menna El Gaafary
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany (M.E.G., B.B., Ta.S., C.Q.S., Th.S.); and Max Planck Institute for Chemical Ecology, Research Group Biosynthesis/NMR, Jena, Germany (S.A., B.S.)
| | - Berthold Büchele
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany (M.E.G., B.B., Ta.S., C.Q.S., Th.S.); and Max Planck Institute for Chemical Ecology, Research Group Biosynthesis/NMR, Jena, Germany (S.A., B.S.)
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany (M.E.G., B.B., Ta.S., C.Q.S., Th.S.); and Max Planck Institute for Chemical Ecology, Research Group Biosynthesis/NMR, Jena, Germany (S.A., B.S.)
| | - Sara Agnolet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany (M.E.G., B.B., Ta.S., C.Q.S., Th.S.); and Max Planck Institute for Chemical Ecology, Research Group Biosynthesis/NMR, Jena, Germany (S.A., B.S.)
| | - Bernd Schneider
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany (M.E.G., B.B., Ta.S., C.Q.S., Th.S.); and Max Planck Institute for Chemical Ecology, Research Group Biosynthesis/NMR, Jena, Germany (S.A., B.S.)
| | - Christoph Q Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany (M.E.G., B.B., Ta.S., C.Q.S., Th.S.); and Max Planck Institute for Chemical Ecology, Research Group Biosynthesis/NMR, Jena, Germany (S.A., B.S.)
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany (M.E.G., B.B., Ta.S., C.Q.S., Th.S.); and Max Planck Institute for Chemical Ecology, Research Group Biosynthesis/NMR, Jena, Germany (S.A., B.S.)
| |
Collapse
|
191
|
Cdk1-mediated phosphorylation of human ATF7 at Thr-51 and Thr-53 promotes cell-cycle progression into M phase. PLoS One 2014; 9:e116048. [PMID: 25545367 PMCID: PMC4278844 DOI: 10.1371/journal.pone.0116048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/30/2014] [Indexed: 12/22/2022] Open
Abstract
Activating transcription factor 2 (ATF2) and its homolog ATF7 are phosphorylated at Thr-69/Thr-71 and at Thr-51/Thr-53, respectively, by stress-activated MAPKs regulating their transcriptional functions in G1 and S phases. However, little is known about the role of ATF2 and ATF7 in G2/M phase. Here, we show that Cdk1-cyclin B1 phosphorylates ATF2 at Thr-69/Thr-71 and ATF7 at Thr-51/Thr-53 from early prophase to anaphase in the absence of any stress stimulation. Knockdown of ATF2 or ATF7 decreases the rate of cell proliferation and the number of cells in M-phase. In particular, the knockdown of ATF7 severely inhibits cell proliferation and G2/M progression. The inducible expression of a mitotically nonphosphorylatable version of ATF7 inhibits G2/M progression despite the presence of endogenous ATF7. We also show that mitotic phosphorylation of ATF7 promotes the activation of Aurora kinases, which are key enzymes for early mitotic events. These results suggest that the Cdk1-mediated phosphorylation of ATF7 facilitates G2/M progression, at least in part, by enabling Aurora signaling.
Collapse
|
192
|
Arnold L, Höckner S, Seufert W. Insights into the cellular mechanism of the yeast ubiquitin ligase APC/C-Cdh1 from the analysis of in vivo degrons. Mol Biol Cell 2014; 26:843-58. [PMID: 25540434 PMCID: PMC4342022 DOI: 10.1091/mbc.e14-09-1342] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The anaphase-promoting complex/cyclosome (APC/C) controls a variety of cellular processes through its ability to target numerous protein substrates for timely degradation. Substrate selection by this ubiquitin ligase depends on related activator proteins, Cdc20 and Cdh1, which bind and activate the APC/C at distinct cell cycle stages. Biochemical and structural studies revealed that Cdc20 and Cdh1 carry conserved receptor domains to recognize specific sequence motifs in substrates, such as D and KEN boxes. The mechanisms for ordered degradation of APC/C substrates, however, remain incompletely understood. Here we describe minimal degradation sequences (degrons) sufficient for rapid APC/C-Cdh1-specific in vivo degradation. The polo kinase Cdc5-derived degron contained an essential KEN motif, whereas a single RxxL-type D box was the relevant signal in the Cdc20-derived degradation domain, indicating that either motif may support specific recognition by Cdh1. In both degrons, the APC/C recognition motif was flanked by a nuclear localization sequence. Forced localization of the degron constructs revealed that proteolysis mediated by APC/C-Cdh1 is restricted to the nucleus and maximally active in the nucleoplasm. Levels of Iqg1, a cytoplasmic Cdh1 substrate, decreased detectably later than the nucleus-localized Cdh1 substrate Ase1, indicating that confinement to the nucleus may allow for temporal control of APC/C-Cdh1-mediated proteolysis.
Collapse
Affiliation(s)
- Lea Arnold
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| | - Sebastian Höckner
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| | - Wolfgang Seufert
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| |
Collapse
|
193
|
Wachowicz P, Chasapi A, Krapp A, Cano Del Rosario E, Schmitter D, Sage D, Unser M, Xenarios I, Rougemont J, Simanis V. Analysis of S. pombe SIN protein association to the SPB reveals two genetically separable states of the SIN. J Cell Sci 2014; 128:741-54. [PMID: 25501816 DOI: 10.1242/jcs.160150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Schizosaccharomyces pombe septation initiation network (SIN) regulates cytokinesis, and asymmetric association of SIN proteins with the mitotic spindle pole bodies (SPBs) is important for its regulation. Here, we have used semi-automated image analysis to study SIN proteins in large numbers of wild-type and mutant cells. Our principal conclusions are: first, that the association of Cdc7p with the SPBs in early mitosis is frequently asymmetric, with a bias in favour of the new SPB; second, that the early association of Cdc7p-GFP to the SPB depends on Plo1p but not Spg1p, and is unaffected by mutations that influence its asymmetry in anaphase; third, that Cdc7p asymmetry in anaphase B is delayed by Pom1p and by activation of the spindle assembly checkpoint, and is promoted by Rad24p; and fourth, that the length of the spindle, expressed as a fraction of the length of the cell, at which Cdc7p becomes asymmetric is similar in cells dividing at different sizes. These data reveal that multiple regulatory mechanisms control the SIN in mitosis and lead us to propose a two-state model to describe the SIN.
Collapse
Affiliation(s)
- Paulina Wachowicz
- Cell cycle control laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), SV-ISREC, 1015 Lausanne, Switzerland
| | - Anastasia Chasapi
- Swiss-Prot. Group and Vital-IT Group, Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Andrea Krapp
- Cell cycle control laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), SV-ISREC, 1015 Lausanne, Switzerland
| | - Elena Cano Del Rosario
- Cell cycle control laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), SV-ISREC, 1015 Lausanne, Switzerland
| | - Daniel Schmitter
- Biomedical Imaging Group, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Daniel Sage
- Biomedical Imaging Group, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Michael Unser
- Biomedical Imaging Group, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Ioannis Xenarios
- Swiss-Prot. Group and Vital-IT Group, Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Jacques Rougemont
- Bioinformatics and Biostatistics Core Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Viesturs Simanis
- Cell cycle control laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), SV-ISREC, 1015 Lausanne, Switzerland
| |
Collapse
|
194
|
Affiliation(s)
- Helder Maiato
- Chromosome Instability & Dynamics Laboratory; Instituto de Biologia Molecular e Celular, Universidade do Porto; Porto Portugal
- Cell Division Unit, Department of Experimental Biology; Faculdade de Medicina, Universidade do Porto; Porto Portugal
| | - Olga Afonso
- Chromosome Instability & Dynamics Laboratory; Instituto de Biologia Molecular e Celular, Universidade do Porto; Porto Portugal
- Cell Division Unit, Department of Experimental Biology; Faculdade de Medicina, Universidade do Porto; Porto Portugal
| | - Irina Matos
- Chromosome Instability & Dynamics Laboratory; Instituto de Biologia Molecular e Celular, Universidade do Porto; Porto Portugal
- Cell Division Unit, Department of Experimental Biology; Faculdade de Medicina, Universidade do Porto; Porto Portugal
| |
Collapse
|
195
|
Eichhorn JM, Kothari A, Chambers TC. Cyclin B1 overexpression induces cell death independent of mitotic arrest. PLoS One 2014; 9:e113283. [PMID: 25415322 PMCID: PMC4240608 DOI: 10.1371/journal.pone.0113283] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/21/2014] [Indexed: 11/23/2022] Open
Abstract
Microtubule inhibitors are widely used in cancer chemotherapy. These drugs characteristically induce mitotic arrest and cell death but the mechanisms linking the two are not firmly established. One of the problems is that cancer cells vary widely in their sensitivity to these agents, and thus comparison of data from different systems is difficult. To alleviate this problem we sought to molecularly induce mitotic death and study its mechanisms, by expressing non-degradable cyclin B (R42A) in HeLa cells. However, this approach failed to induce significant mitotic arrest, Cdk1 activation, or phosphorylation of anti-apoptotic Bcl-2 proteins, all characteristics of cells treated with microtubule inhibitors. Furthermore, cyclin B1-R42A induced rapid cell death, and when expressed in synchronized cells, cell death occurred in G1 phase. Decreasing the plasmid concentration reduced transfection efficiency but restored mitotic arrest and eliminated non-specific death. These results show that inappropriate overexpression of cyclin B1 causes non-specific cell death and suggest caution in its use for the study of mitotic events.
Collapse
Affiliation(s)
- Joshua M. Eichhorn
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arizona, United States of America
| | - Anisha Kothari
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arizona, United States of America
| | - Timothy C. Chambers
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arizona, United States of America
| |
Collapse
|
196
|
Muro E, Gébrane-Younès J, Jobart-Malfait A, Louvet E, Roussel P, Hernandez-Verdun D. The traffic of proteins between nucleolar organizer regions and prenucleolar bodies governs the assembly of the nucleolus at exit of mitosis. Nucleus 2014. [DOI: 10.4161/nucl.11334] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
197
|
London N, Biggins S. Signalling dynamics in the spindle checkpoint response. Nat Rev Mol Cell Biol 2014; 15:736-47. [PMID: 25303117 DOI: 10.1038/nrm3888] [Citation(s) in RCA: 245] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The spindle checkpoint ensures proper chromosome segregation during cell division. Unravelling checkpoint signalling has been a long-standing challenge owing to the complexity of the structures and forces that regulate chromosome segregation. New reports have now substantially advanced our understanding of checkpoint signalling mechanisms at the kinetochore, the structure that connects microtubules and chromatin. In contrast to the traditional view of a binary checkpoint response - either completely on or off - new findings indicate that the checkpoint response strength is variable. This revised perspective provides insight into how checkpoint bypass can lead to aneuploidy and informs strategies to exploit these errors for cancer treatments.
Collapse
Affiliation(s)
- Nitobe London
- 1] Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N., PO Box 19024, Seattle, Washington 98109, USA. [2] Molecular and Cellular Biology Program, University of Washington/Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Sue Biggins
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N., PO Box 19024, Seattle, Washington 98109, USA
| |
Collapse
|
198
|
Lu D, Hsiao JY, Davey NE, Van Voorhis VA, Foster SA, Tang C, Morgan DO. Multiple mechanisms determine the order of APC/C substrate degradation in mitosis. ACTA ACUST UNITED AC 2014; 207:23-39. [PMID: 25287299 PMCID: PMC4195823 DOI: 10.1083/jcb.201402041] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
To ensure proper mitotic progression, robust ordering of the destruction of APC/CCdc20 substrates is driven by the integration of molecular mechanisms ranging from phosphorylation-dependent interaction with substrates to sensing of the status of the spindle assembly checkpoint. The ubiquitin protein ligase anaphase-promoting complex or cyclosome (APC/C) controls mitosis by promoting ordered degradation of securin, cyclins, and other proteins. The mechanisms underlying the timing of APC/C substrate degradation are poorly understood. We explored these mechanisms using quantitative fluorescence microscopy of GFP-tagged APC/CCdc20 substrates in living budding yeast cells. Degradation of the S cyclin, Clb5, begins early in mitosis, followed 6 min later by the degradation of securin and Dbf4. Anaphase begins when less than half of securin is degraded. The spindle assembly checkpoint delays the onset of Clb5 degradation but does not influence securin degradation. Early Clb5 degradation depends on its interaction with the Cdk1–Cks1 complex and the presence of a Cdc20-binding “ABBA motif” in its N-terminal region. The degradation of securin and Dbf4 is delayed by Cdk1-dependent phosphorylation near their Cdc20-binding sites. Thus, a remarkably diverse array of mechanisms generates robust ordering of APC/CCdc20 substrate destruction.
Collapse
Affiliation(s)
- Dan Lu
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Jennifer Y Hsiao
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Norman E Davey
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Vanessa A Van Voorhis
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Scott A Foster
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Chao Tang
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - David O Morgan
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| |
Collapse
|
199
|
Chen J, Liu J. Spatial-temporal model for silencing of the mitotic spindle assembly checkpoint. Nat Commun 2014; 5:4795. [PMID: 25216458 PMCID: PMC4163959 DOI: 10.1038/ncomms5795] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 07/24/2014] [Indexed: 01/07/2023] Open
Abstract
The spindle assembly checkpoint arrests mitotic progression until each kinetochore secures a stable attachment to the spindle. Despite fluctuating noise, this checkpoint remains robust and remarkably sensitive to even a single unattached kinetochore among many attached kinetochores; moreover, the checkpoint is silenced only after the final kinetochore-spindle attachment. Experimental observations have shown that checkpoint components stream from attached kinetochores along microtubules toward spindle poles. Here, we incorporate this streaming behavior into a theoretical model that accounts for the robustness of checkpoint silencing. Poleward streams are integrated at spindle poles, but are diverted by any unattached kinetochore; consequently, accumulation of checkpoint components at spindle poles increases markedly only when every kinetochore is properly attached. This step-change robustly triggers checkpoint silencing after, and only after, the final kinetochore-spindle attachment. Our model offers a conceptual framework that highlights the role of spatiotemporal regulation in mitotic spindle checkpoint signaling and fidelity of chromosome segregation.
Collapse
Affiliation(s)
- Jing Chen
- National Heart, Lung and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Room 3306, Bethesda, Maryland 20892, USA
| | - Jian Liu
- National Heart, Lung and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Room 3306, Bethesda, Maryland 20892, USA
| |
Collapse
|
200
|
Matsusaka T, Enquist-Newman M, Morgan DO, Pines J. Co-activator independent differences in how the metaphase and anaphase APC/C recognise the same substrate. Biol Open 2014; 3:904-12. [PMID: 25217616 PMCID: PMC4197439 DOI: 10.1242/bio.20149415] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/13/2014] [Indexed: 12/20/2022] Open
Abstract
The Anaphase Promoting Complex or Cyclosome (APC/C) is critical to the control of mitosis. The APC/C is an ubiquitin ligase that targets specific mitotic regulators for proteolysis at distinct times in mitosis, but how this is achieved is not well understood. We have addressed this question by determining whether the same substrate, cyclin B1, is recognised in the same way by the APC/C at different times in mitosis. Unexpectedly, we find that distinct but overlapping motifs in cyclin B1 are recognised by the APC/C in metaphase compared with anaphase, and this does not depend on the exchange of Cdc20 for Cdh1. Thus, changes in APC/C substrate specificity in mitosis can potentially be conferred by altering interaction sites in addition to exchanging Cdc20 for Cdh1.
Collapse
Affiliation(s)
- Takahiro Matsusaka
- The Gurdon Institute, Tennis Court Road, Cambridge CB2 1QN, UK Department of Zoology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Maria Enquist-Newman
- Department of Physiology, University of California in San Francisco (UCSF), 600 16th Street, San Francisco, CA 94158, USA
| | - David O Morgan
- Department of Physiology, University of California in San Francisco (UCSF), 600 16th Street, San Francisco, CA 94158, USA
| | - Jonathon Pines
- The Gurdon Institute, Tennis Court Road, Cambridge CB2 1QN, UK Department of Zoology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| |
Collapse
|