1
|
Brigant B, Demont Y, Ouled-Haddou H, Metzinger-Le Meuth V, Testelin S, Garçon L, Metzinger L, Rochette J. TRIM37 is highly expressed during mitosis in CHON-002 chondrocytes cell line and is regulated by miR-223. Bone 2020; 137:115393. [PMID: 32353567 DOI: 10.1016/j.bone.2020.115393] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023]
Abstract
Multiple molecular disorders can affect mechanisms regulating proliferation and differentiation of growth plate chondrocytes. Mutations in the TRIM37 gene cause the Mulibrey nanism, a heritable growth disorder. Since chondrocytes are instrumental in long bone growth that is deficient in nanism, we hypothesized that TRIM37 defect could contribute to dysregulation of the chondrocyte cell cycle. Western blotting, confocal microscopy and imaging flow cytometry determined TRIM37 expression in CHON-002 cell lineage. We showed that TRIM37 is expressed during mitosis of chondrocytes and directly impacted their proliferation. During the chondrocyte cell cycle, TRIM37 was present in both nucleus and cytoplasm. During M phase we observed an increase of the TRIM37-Tubulin co-localization in comparison with G1, S and G2 phases. TRIM37 knock down inhibited proliferation, together with cell cycle anomalies and increased autophagy, while overexpression accordingly enhanced cell proliferation. We demonstrated that microRNA-223 directly targets TRIM37, and suggest that miR-223 regulates TRIM37 gene expression during the cell cycle. In summary, our results give clues to explain why TRIM37 deficiency in chondrocytes impacts bone growth. Modulating TRIM37 using miR-223 could be an approach to increase chondrogenesis.
Collapse
Affiliation(s)
- Benjamin Brigant
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Yohann Demont
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Hakim Ouled-Haddou
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | | | - Sylvie Testelin
- Maxillo-Facial Surgery Department, Centre Hospitalo-Universitaire d'Amiens, Avenue Laennec, 80000 Amiens, France; EA CHIMERE, université de Picardie-Jules-Verne, Avenue Laennec, 80000 Amiens, France; Facing Faces Institute, Avenue Laennec, 80000 Amiens, France
| | - Loïc Garçon
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Laurent Metzinger
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Jacques Rochette
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France.
| |
Collapse
|
2
|
Liu F, Fan Y, Ou L, Li T, Fan J, Duan L, Yang J, Luo C, Wu X. CircHIPK3 Facilitates the G2/M Transition in Prostate Cancer Cells by Sponging miR-338-3p. Onco Targets Ther 2020; 13:4545-4558. [PMID: 32547085 PMCID: PMC7251229 DOI: 10.2147/ott.s242482] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 05/01/2020] [Indexed: 01/13/2023] Open
Abstract
Background Circular RNAs (circRNAs) play a crucial role in gene expression regulation. CircHIPK3 is a circRNA derived from Exon 2 of HIPK3 gene and its role in prostate cancer (PCa) is still unclear. Methods CCK8 assays, flow cytometry and colony formation assays were performed to assess the effects of circHIPK3 in PCa cells. Bioinformatics analysis, RNA pull-down assay, RNA immunoprecipitation assay (RIP), and luciferase activity assay were performed to dissect the mechanism underlying circHIPK3-mediated G2/M transition in PCa cells. Results CircHIPK3 expression was upregulated in PCa cells and prostate cancer tissues. Overexpression of circHIPK3 or circHIPK3 silencing altered PCa viability, proliferation and apoptosis in vitro. CircHIPK3 could sponge miR-338-3p and inhibit its activity, resulting in increased expression of Cdc25B and Cdc2 in vitro. Conclusion CircHIPK3 promotes G2/M transition and induces PCa cell proliferation by sponging miR-338-3p and increasing the expression of Cdc25B and Cdc2. CircHIPK3 may play an oncogenic role in PCa.
Collapse
Affiliation(s)
- Fengchun Liu
- Department of Laboratory Diagnosis, Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| | - Yanru Fan
- Department of Laboratory Diagnosis, Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| | - Liping Ou
- Department of Laboratory Diagnosis, Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| | - Ting Li
- Department of Laboratory Diagnosis, Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| | - Jiaxin Fan
- Department of Laboratory Diagnosis, Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| | - Limei Duan
- Department of Laboratory Diagnosis, Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| | - Jinxiao Yang
- Department of Laboratory Diagnosis, Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| | - Chunli Luo
- Department of Laboratory Diagnosis, Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| | - Xiaohou Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 408000, People's Republic of China
| |
Collapse
|
3
|
Lam CW, Fong NC, Chan TYC, Lau KC, Ling TK, Mak DWY, Cheng X, Law CY. Centrosome-associated CDC25B is a novel disease-causing gene for a syndrome with cataracts, dilated cardiomyopathy, and multiple endocrinopathies. Clin Chim Acta 2020; 504:81-87. [PMID: 32027886 DOI: 10.1016/j.cca.2020.01.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/07/2020] [Accepted: 01/17/2020] [Indexed: 11/16/2022]
Abstract
We describe a unique Chinese girl who presented with intrauterine growth retardation, delayed development, bilateral cataracts, hypothyroidism, growth hormone deficiency, and juvenile dilated cardiomyopathy. She was born to consanguineous parents with a history of one fetal and one infantile death in the family. She died from cardiac failure at the age of 12. In the pursuit of a diagnosis, the family was referred to the Clinics for Rare Diseases Referral and the University of Hong Kong Undiagnosed Disease Program. Whole-exome sequencing analysis revealed a homozygous non-sense variant, NM_021873:c.313G > T (p.Glu105*), in the CDC25B gene, a key regulator of the cell cycle. This variant was located in a region of homozygosity of 25 Mb on chromosome 20. Her parents and two asymptomatic sisters were confirmed to be carriers and one brother did not carry the variant. This is the first report of a natural human knockout of the CDC25B gene. Multiple endocrinopathies and fatal juvenile dilated cardiomyopathy suggests the potential for unfavorable complications in oncology patients receiving CDC25B inhibitors as an emerging targeted therapy.
Collapse
Affiliation(s)
- Ching-Wan Lam
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.
| | - Nai-Chung Fong
- Department of Paediatrics & Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China
| | | | - Kwai-Cheung Lau
- Department of Pathology, Princess Margaret Hospital, Hong Kong, China
| | - Tsz-Ki Ling
- Division of Chemical Pathology, Department of Pathology, Queen Mary Hospital, Hong Kong, China
| | - Daniel Wai-Yau Mak
- Department of Paediatrics & Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China
| | - Xinqi Cheng
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chun-Yiu Law
- Division of Chemical Pathology, Department of Pathology, Queen Mary Hospital, Hong Kong, China
| |
Collapse
|
4
|
Crncec A, Hochegger H. Triggering mitosis. FEBS Lett 2019; 593:2868-2888. [PMID: 31602636 DOI: 10.1002/1873-3468.13635] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/07/2019] [Accepted: 10/07/2019] [Indexed: 12/28/2022]
Abstract
Entry into mitosis is triggered by the activation of cyclin-dependent kinase 1 (Cdk1). This simple reaction rapidly and irreversibly sets the cell up for division. Even though the core step in triggering mitosis is so simple, the regulation of this cellular switch is highly complex, involving a large number of interconnected signalling cascades. We do have a detailed knowledge of most of the components of this network, but only a poor understanding of how they work together to create a precise and robust system that ensures that mitosis is triggered at the right time and in an orderly fashion. In this review, we will give an overview of the literature that describes the Cdk1 activation network and then address questions relating to the systems biology of this switch. How is the timing of the trigger controlled? How is mitosis insulated from interphase? What determines the sequence of events, following the initial trigger of Cdk1 activation? Which elements ensure robustness in the timing and execution of the switch? How has this system been adapted to the high levels of replication stress in cancer cells?
Collapse
Affiliation(s)
- Adrijana Crncec
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| | - Helfrid Hochegger
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| |
Collapse
|
5
|
Holmes TR, Dindu S, Hansen LA. Aberrant localization of signaling proteins in skin cancer: Implications for treatment. Mol Carcinog 2019; 58:1631-1639. [PMID: 31062427 DOI: 10.1002/mc.23036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 01/01/2023]
Abstract
Aberrant subcellular localization of signaling proteins can provide cancer cells with advantages such as resistance to apoptotic cell death, increased invasiveness and more rapid proliferation. Nuclear to cytoplasmic shifts in tumor-promoting proteins can lead to worse patient outcomes, providing opportunities to target cancer-specific processes. Herein, we review the significance of dysregulated protein localization with a focus on skin cancer. Altered localization of signaling proteins controlling cell cycle progression or cell death is a common feature of cancer. In some instances, aberrant subcellular localization results in an acquired prosurvival function. Taking advantage of this knowledge reveals novel targets useful in the development of cancer therapeutics.
Collapse
Affiliation(s)
- Thomas R Holmes
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Shravya Dindu
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Laura A Hansen
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| |
Collapse
|
6
|
Al‐Matouq J, Holmes TR, Hansen LA. CDC25B and CDC25C overexpression in nonmelanoma skin cancer suppresses cell death. Mol Carcinog 2019; 58:1691-1700. [DOI: 10.1002/mc.23075] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/29/2019] [Accepted: 06/02/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Jenan Al‐Matouq
- Department of Biomedical SciencesCreighton University Omaha Nebraska
| | - Thomas R. Holmes
- Department of Biomedical SciencesCreighton University Omaha Nebraska
| | - Laura A. Hansen
- Department of Biomedical SciencesCreighton University Omaha Nebraska
| |
Collapse
|
7
|
Pintard L, Archambault V. A unified view of spatio-temporal control of mitotic entry: Polo kinase as the key. Open Biol 2018; 8:180114. [PMID: 30135239 PMCID: PMC6119860 DOI: 10.1098/rsob.180114] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 07/29/2018] [Indexed: 12/18/2022] Open
Abstract
The Polo kinase is an essential regulator of cell division. Its ability to regulate multiple events at distinct subcellular locations and times during mitosis is remarkable. In the last few years, a much clearer mechanistic understanding of the functions and regulation of Polo in cell division has emerged. In this regard, the importance of coupling changes in activity with changes in localization is striking, both for Polo itself and for its upstream regulators. This review brings together several new pieces of the puzzle that are gradually revealing how Polo is regulated, in space and time, to enable its functions in the early stages of mitosis in animal cells. As a result, a unified view of how mitotic entry is spatio-temporally regulated is emerging.
Collapse
Affiliation(s)
- Lionel Pintard
- Cell Cycle and Development Team, Institut Jacques Monod, UMR7592 CNRS-Université Paris Diderot, Sorbonne Paris Cité, Ligue contre le Cancer, Paris, France
- Equipe labellisée, Ligue contre le Cancer, Paris, France
| | - Vincent Archambault
- Institut de recherche en immunologie et en cancérologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
8
|
MASTL overexpression promotes chromosome instability and metastasis in breast cancer. Oncogene 2018; 37:4518-4533. [PMID: 29743597 PMCID: PMC6095835 DOI: 10.1038/s41388-018-0295-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/27/2018] [Accepted: 04/08/2018] [Indexed: 12/13/2022]
Abstract
MASTL kinase is essential for correct progression through mitosis, with loss of MASTL causing chromosome segregation errors, mitotic collapse and failure of cytokinesis. However, in cancer MASTL is most commonly amplified and overexpressed. This correlates with increased chromosome instability in breast cancer and poor patient survival in breast, ovarian and lung cancer. Global phosphoproteomic analysis of immortalised breast MCF10A cells engineered to overexpressed MASTL revealed disruption to desmosomes, actin cytoskeleton, PI3K/AKT/mTOR and p38 stress kinase signalling pathways. Notably, these pathways were also disrupted in patient samples that overexpress MASTL. In MCF10A cells, these alterations corresponded with a loss of contact inhibition and partial epithelial-mesenchymal transition, which disrupted migration and allowed cells to proliferate uncontrollably in 3D culture. Furthermore, MASTL overexpression increased aberrant mitotic divisions resulting in increased micronuclei formation. Mathematical modelling indicated that this delay was due to continued inhibition of PP2A-B55, which delayed timely mitotic exit. This corresponded with an increase in DNA damage and delayed transit through interphase. There were no significant alterations to replication kinetics upon MASTL overexpression, however, inhibition of p38 kinase rescued the interphase delay, suggesting the delay was a G2 DNA damage checkpoint response. Importantly, knockdown of MASTL, reduced cell proliferation, prevented invasion and metastasis of MDA-MB-231 breast cancer cells both in vitro and in vivo, indicating the potential of future therapies that target MASTL. Taken together, these results suggest that MASTL overexpression contributes to chromosome instability and metastasis, thereby decreasing breast cancer patient survival.
Collapse
|
9
|
Kachaner D, Garrido D, Mehsen H, Normandin K, Lavoie H, Archambault V. Coupling of Polo kinase activation to nuclear localization by a bifunctional NLS is required during mitotic entry. Nat Commun 2017; 8:1701. [PMID: 29167465 PMCID: PMC5700101 DOI: 10.1038/s41467-017-01876-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/22/2017] [Indexed: 02/07/2023] Open
Abstract
The Polo kinase is a master regulator of mitosis and cytokinesis conserved from yeasts to humans. Polo is composed of an N-term kinase domain (KD) and a C-term polo-box domain (PBD), which regulates its subcellular localizations. The PBD and KD can interact and inhibit each other, and this reciprocal inhibition is relieved when Polo is phosphorylated at its activation loop. How Polo activation and localization are coupled during mitotic entry is unknown. Here we report that PBD binding to the KD masks a nuclear localization signal (NLS). Activating phosphorylation of the KD leads to exposure of the NLS and entry of Polo into the nucleus before nuclear envelope breakdown. Failures of this mechanism result in misregulation of the Cdk1-activating Cdc25 phosphatase and lead to mitotic and developmental defects in Drosophila. These results uncover spatiotemporal mechanisms linking master regulatory enzymes during mitotic entry. Drosophila Polo kinase is the founding member of the Polo-Like Kinase (PLK) family and a master regulator of mitosis and cytokinesis. Here the authors uncover a molecular mechanism for the spatiotemporal regulation of Polo kinase during mitotic entry through a phosphorylation event that triggers nuclear import.
Collapse
Affiliation(s)
- David Kachaner
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7.,Département de biochimie et médecine moléculaire, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7
| | - Damien Garrido
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7.,Département de biochimie et médecine moléculaire, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7
| | - Haytham Mehsen
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7
| | - Karine Normandin
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7
| | - Hugo Lavoie
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7
| | - Vincent Archambault
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7. .,Département de biochimie et médecine moléculaire, Université de Montréal, C.P. 6128 Succursale Centre-Ville, Montréal, QC, Canada, H3C 3J7.
| |
Collapse
|
10
|
Cui C, Zang T, Cao Y, Qin X, Zhang X. CDC25B is involved in the centrosomal microtubule nucleation in two-cell stage mouse embryos. Dev Growth Differ 2016; 58:714-726. [PMID: 27885657 DOI: 10.1111/dgd.12328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 01/21/2023]
Abstract
CDC25B has been demonstrated to activate the complex of CDK1/Cyclin B and trigger mitosis. We have recently demonstrated that p-CDC25B-Ser351 is located at the centrosomes of mouse oocytes and contributes to the release of mouse oocytes from prophase I arrest. But much less is known about CDC25B function at the centrosome in two-cell stage mouse embryos. Here we investigate the effect of CDC25B regulating the microtubules nucleation. Microinjection of anti-CDC25B antibody caused aberrant microtubule nucleation. In addition, embryos injected with anti-CDC25B antibody showed the marked absence of microtubule repolymerization and Nek2 foci after nocodazole washout. CDC25B overexpression caused microtubule-organizing center (MTOC) overduplication. Moreover, overexpression of CDC25B-▵65 mutant resulted in the loss of CDC25B localization in the perinuclear region and made CDC25B less efficient in inducing mitosis. We additionally identified that CDC25B is responsible for the pericentrin localization to the MTOC. Our data suggest an important role of CDC25B for microtubule nucleation and organization. N-terminal of CDC25B is required for regulating the microtubule dynamics and mitotic function.
Collapse
Affiliation(s)
- Cheng Cui
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Tianxia Zang
- Department of Endocrinology, The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110034, Liaoning, China
| | - Yu Cao
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xin Qin
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xuewei Zhang
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| |
Collapse
|
11
|
14-3-3γ Prevents Centrosome Amplification and Neoplastic Progression. Sci Rep 2016; 6:26580. [PMID: 27253419 PMCID: PMC4890593 DOI: 10.1038/srep26580] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/04/2016] [Indexed: 12/21/2022] Open
Abstract
More than 80% of malignant tumors show centrosome amplification and clustering. Centrosome amplification results from aberrations in the centrosome duplication cycle, which is strictly coordinated with DNA-replication-cycle. However, the relationship between cell-cycle regulators and centrosome duplicating factors is not well understood. This report demonstrates that 14-3-3γ localizes to the centrosome and 14-3-3γ loss leads to centrosome amplification. Loss of 14-3-3γ results in the phosphorylation of NPM1 at Thr-199, causing early centriole disjunction and centrosome hyper-duplication. The centrosome amplification led to aneuploidy and increased tumor formation in mice. Importantly, an increase in passage of the 14-3-3γ-knockdown cells led to an increase in the number of cells containing clustered centrosomes leading to the generation of pseudo-bipolar spindles. The increase in pseudo-bipolar spindles was reversed and an increase in the number of multi-polar spindles was observed upon expression of a constitutively active 14-3-3-binding-defective-mutant of cdc25C (S216A) in the 14-3-3γ knockdown cells. The increase in multi-polar spindle formation was associated with decreased cell viability and a decrease in tumor growth. Our findings uncover the molecular basis of regulation of centrosome duplication by 14-3-3γ and inhibition of tumor growth by premature activation of the mitotic program and the disruption of centrosome clustering.
Collapse
|
12
|
Phosphatases and kinases regulating CDC25 activity in the cell cycle: clinical implications of CDC25 overexpression and potential treatment strategies. Mol Cell Biochem 2016; 416:33-46. [PMID: 27038604 DOI: 10.1007/s11010-016-2693-2] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 03/24/2016] [Indexed: 10/22/2022]
Abstract
Alterations in the cell-cycle regulatory genes result in uncontrolled cell proliferation leading to several disease conditions. Cyclin-dependent kinases (CDK) and their regulatory subunit, cyclins, are essential proteins in cell-cycle progression. The activity of CDK is regulated by a series of phosphorylation and dephosphorylation at different amino acid residues. Cell Division Cycle-25 (CDC25) plays an important role in transitions between cell-cycle phases by dephosphorylating and activating CDKs. CDC25B and CDC25C play a major role in G2/M progression, whereas CDC25A assists in G1/S transition. Different isomers of CDC25 expressions are upregulated in various clinicopathological situations. Overexpression of CDC25A deregulates G1/S and G2/M events, including the G2 checkpoint. CDC25B has oncogenic properties. Binding to the 14-3-3 proteins regulates the activity and localization of CDC25B. CDC25C is predominantly a nuclear protein in mammalian cells. At the G2/M transition, mitotic activation of CDC25C protein occurs by its dissociation from 14-3-3 proteins along with its phosphorylation at multiple sites within its N-terminal domain. In this article, we critically reviewed the biology of the activation/deactivation of CDC25 by kinases/phosphatases to maintain the level of CDK-cyclin activities and thus the genomic stability, clinical implications due to dysregulation of CDC25, and potential role of CDC25 inhibitors in diseases.
Collapse
|
13
|
Garcia PL, Miller AL, Kreitzburg KM, Council LN, Gamblin TL, Christein JD, Heslin MJ, Arnoletti JP, Richardson JH, Chen D, Hanna CA, Cramer SL, Yang ES, Qi J, Bradner JE, Yoon KJ. The BET bromodomain inhibitor JQ1 suppresses growth of pancreatic ductal adenocarcinoma in patient-derived xenograft models. Oncogene 2015; 35:833-45. [PMID: 25961927 DOI: 10.1038/onc.2015.126] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/04/2015] [Accepted: 03/16/2015] [Indexed: 12/23/2022]
Abstract
The primary aim of this study was to evaluate the antitumor efficacy of the bromodomain inhibitor JQ1 in pancreatic ductal adenocarcinoma (PDAC) patient-derived xenograft (tumorgraft) models. A secondary aim of the study was to evaluate whether JQ1 decreases expression of the oncogene c-Myc in PDAC tumors, as has been reported for other tumor types. We used five PDAC tumorgraft models that retain specific characteristics of tumors of origin to evaluate the antitumor efficacy of JQ1. Tumor-bearing mice were treated with JQ1 (50 mg/kg daily for 21 or 28 days). Expression analyses were performed with tumors harvested from host mice after treatment with JQ1 or vehicle control. An nCounter PanCancer Pathways Panel (NanoString Technologies) of 230 cancer-related genes was used to identify gene products affected by JQ1. Quantitative RT-PCR, immunohistochemistry and immunoblots were carried out to confirm that changes in RNA expression reflected changes in protein expression. JQ1 inhibited the growth of all five tumorgraft models (P<0.05), each of which harbors a KRAS mutation; but induced no consistent change in expression of c-Myc protein. Expression profiling identified CDC25B, a regulator of cell cycle progression, as one of the three RNA species (TIMP3, LMO2 and CDC25B) downregulated by JQ1 (P<0.05). Inhibition of tumor progression was more closely related to decreased expression of nuclear CDC25B than to changes in c-Myc expression. JQ1 and other agents that inhibit the function of proteins with bromodomains merit further investigation for treating PDAC tumors. Work is ongoing in our laboratory to identify effective drug combinations that include JQ1.
Collapse
Affiliation(s)
- P L Garcia
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - A L Miller
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - K M Kreitzburg
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - L N Council
- Division of Anatomic Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - T L Gamblin
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J D Christein
- Division of General Surgery, Gastrointestinal Surgery or Surgical Oncology, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M J Heslin
- Division of General Surgery, Gastrointestinal Surgery or Surgical Oncology, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J P Arnoletti
- Division of General Surgery, Gastrointestinal Surgery or Surgical Oncology, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J H Richardson
- Division of General Surgery, Gastrointestinal Surgery or Surgical Oncology, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - D Chen
- Division of Preventive Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - C A Hanna
- Division of Anatomic Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - S L Cramer
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - E S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Qi
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - J E Bradner
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - K J Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
14
|
Wang M, Zhu XY, Wang L, Lin Y. Expression and significance of CDC25B, PED/PEA-15 in esophageal carcinoma. Cancer Biother Radiopharm 2015; 30:139-45. [PMID: 25775393 DOI: 10.1089/cbr.2014.1701] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE To explore the role of CDC25B, PED/PEA-15 in the development of esophageal carcinoma and its influence on the prognosis. METHODS Fluorescence quantitative real-time PCR and immunohistochemistry methods were used to analyze the expression of CDC25B, PED/PEA-15 in esophageal carcinoma. Moreover, survival analysis was done using the Kaplan-Meier method. RESULTS In 66 cases of esophageal cancer tissues, the relative content of CDC25B mRNA was 16.22 (13.93-18.90). The positive expression rate of CDC25B protein was 48.5%, significantly higher than normal mucosa tissues (0%) (p<0.01). The relative content of PED/PEA-15 mRNA was 12.47 (10.41-14.93). The positive expression rate of PED/PEA-15 protein was 68.2%, significantly higher than normal mucosa tissues (17.6%) (p<0.01). The CDC25B protein expression was correlated with differentiation grade and depth of invasion (p<0.05). The PED/PEA-15 protein expression was related to differentiation grade, lymph node metastasis, and depth of invasion (p<0.05). Survival analysis showed that the mean survival time of PED/PEA-15-positive expression group was lower compared with the negative expression group (χ(2)=5.549, p=0.018). Analysis of the relationship between CDC25B and PED/PEA-15 suggested that there was a positive correlation between them (r=4.061, p=0.044). CONCLUSION Both CDC25B and PED/PEA-15 play a certain role in the carcinogenesis of esophageal cancer, and PED/PEA-15 has a greater influence on postoperative survival time. They will be the new diagnostic/therapeutic targets in esophageal carcinoma.
Collapse
Affiliation(s)
- Ming Wang
- 1 Department of Radiation Therapy, Cangzhou Central Hospital , Cangzhou, China
| | | | | | | |
Collapse
|
15
|
Zarling AL, Obeng RC, Desch AN, Pinczewski J, Cummings KL, Deacon DH, Conaway M, Slingluff CL, Engelhard VH. MHC-restricted phosphopeptides from insulin receptor substrate-2 and CDC25b offer broad-based immunotherapeutic agents for cancer. Cancer Res 2014; 74:6784-95. [PMID: 25297629 DOI: 10.1158/0008-5472.can-14-0043] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cancer cells display novel phosphopeptides in association with MHC class I and II molecules. In this study, we evaluated two HLA-A2-restricted phosphopeptides derived from the insulin receptor substrate (IRS)-2 and the cell-cycle regulator CDC25b. These proteins are both broadly expressed in multiple malignancies and linked to cancer cell survival. Two phosphopeptides, termed pIRS-21097-1105 and pCDC25b38-46, served as targets of strong and specific CD8 T-cell memory responses in normal human donors. We cloned T-cell receptor (TCR) cDNAs from murine CD8 T-cell lines specific for either pIRS-21097-1105 or pCDC25b38-46. Expression of these TCRs in human CD8 T cells imparted high-avidity phosphopeptide-specific recognition and cytotoxic and cytokine-secreting effector activities. Using these cells, we found that endogenously processed pIRS-21097-1105 was presented on HLA-A2(+) melanomas and breast, ovarian, and colorectal carcinomas. Presentation was correlated with the level of the Ser(1100)-phosphorylated IRS-2 protein in metastatic melanoma tissues. The highest expression of this protein was evident on dividing malignant cells. Presentation of endogenously processed pCDC25b38-46 was narrower, but still evident on HLA-A2(+) melanoma, breast carcinoma, and lymphoblastoid cells. Notably, pIRS-21097-1105-specific and pCDC25b38-46-specific TCR-expressing human CD8 T cells markedly slowed tumor outgrowth in vivo. Our results define two new antigens that may be developed as immunotherapeutic agents for a broad range of HLA-A2(+) cancers.
Collapse
Affiliation(s)
- Angela L Zarling
- Carter Immunology Center and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Rebecca C Obeng
- Carter Immunology Center and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - A Nicole Desch
- Carter Immunology Center and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Joel Pinczewski
- Department of Pathology, University of Virginia, Charlottesville, Virginia. Human Immune Therapy Center, University of Virginia, Charlottesville, Virginia
| | - Kara L Cummings
- Carter Immunology Center and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Donna H Deacon
- Human Immune Therapy Center, University of Virginia, Charlottesville, Virginia. Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Mark Conaway
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
| | - Craig L Slingluff
- Human Immune Therapy Center, University of Virginia, Charlottesville, Virginia. Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Victor H Engelhard
- Carter Immunology Center and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
16
|
Cui C, Ren X, Liu D, Deng X, Qin X, Zhao X, Wang E, Yu B. 14-3-3 epsilon prevents G2/M transition of fertilized mouse eggs by binding with CDC25B. BMC DEVELOPMENTAL BIOLOGY 2014; 14:33. [PMID: 25059436 PMCID: PMC4222595 DOI: 10.1186/s12861-014-0033-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 07/14/2014] [Indexed: 12/21/2022]
Abstract
Background The 14-3-3 (YWHA) proteins are highly conserved in higher eukaryotes, participate in various cellular signaling pathways including cell cycle regulation, development and growth. Our previous studies demonstrated that 14-3-3ε (YWHAE) is responsible for maintaining prophase | arrest in mouse oocyte. However, roles of 14-3-3ε in the mitosis of fertilized mouse eggs have remained unclear. Here, we showed that 14-3-3ε interacts and cooperates with CDC25B phosphorylated at Ser321 regulating G2/M transition of mitotic progress of fertilized mouse eggs. Results Disruption of 14-3-3ε expression by RNAi prevented normal G2/M transition by inhibition of MPF activity and leaded to the translocation of CDC25B into the nucleus from the cytoplasm. Overexpression of 14-3-3ε-WT and unphosphorylatable CDC25B mutant (CDC25B-S321A) induced mitotic resumption in dbcAMP-arrested eggs. In addition, we examined endogenous and exogenous distribution of 14-3-3ε and CDC25B. Endogenous 14-3-3ε and CDC25B were co-localized primarily in the cytoplasm at the G1, S, early G2 and M phases whereas CDC25B was found to accumulate in the nucleus at the late G2 phase. Upon coexpression with RFP–14-3-3ε, GFP–CDC25B–WT and GFP–CDC25B–S321A were predominantly cytoplasmic at early G2 phase and then GFP–CDC25B–S321A moved to the nucleus whereas CDC25B-WT signals were observed in the cytoplasm without nucleus accumulation at late G2 phase at presence of dbcAMP. Conclusions Our data indicate that 14-3-3ε is required for the mitotic entry in the fertilized mouse eggs. 14-3-3ε is primarily responsible for sequestering the CDC25B in cytoplasm and 14-3-3ε binding to CDC25B-S321 phosphorylated by PKA induces mitotic arrest at one-cell stage by inactivation of MPF in fertilized mouse eggs.
Collapse
|
17
|
Ali Hassan NZ, Mokhtar NM, Kok Sin T, Mohamed Rose I, Sagap I, Harun R, Jamal R. Integrated analysis of copy number variation and genome-wide expression profiling in colorectal cancer tissues. PLoS One 2014; 9:e92553. [PMID: 24694993 PMCID: PMC3973632 DOI: 10.1371/journal.pone.0092553] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 02/24/2014] [Indexed: 12/24/2022] Open
Abstract
Integrative analyses of multiple genomic datasets for selected samples can provide better insight into the overall data and can enhance our knowledge of cancer. The objective of this study was to elucidate the association between copy number variation (CNV) and gene expression in colorectal cancer (CRC) samples and their corresponding non-cancerous tissues. Sixty-four paired CRC samples from the same patients were subjected to CNV profiling using the Illumina HumanOmni1-Quad assay, and validation was performed using multiplex ligation probe amplification method. Genome-wide expression profiling was performed on 15 paired samples from the same group of patients using the Affymetrix Human Gene 1.0 ST array. Significant genes obtained from both array results were then overlapped. To identify molecular pathways, the data were mapped to the KEGG database. Whole genome CNV analysis that compared primary tumor and non-cancerous epithelium revealed gains in 1638 genes and losses in 36 genes. Significant gains were mostly found in chromosome 20 at position 20q12 with a frequency of 45.31% in tumor samples. Examples of genes that were associated at this cytoband were PTPRT, EMILIN3 and CHD6. The highest number of losses was detected at chromosome 8, position 8p23.2 with 17.19% occurrence in all tumor samples. Among the genes found at this cytoband were CSMD1 and DLC1. Genome-wide expression profiling showed 709 genes to be up-regulated and 699 genes to be down-regulated in CRC compared to non-cancerous samples. Integration of these two datasets identified 56 overlapping genes, which were located in chromosomes 8, 20 and 22. MLPA confirmed that the CRC samples had the highest gains in chromosome 20 compared to the reference samples. Interpretation of the CNV data in the context of the transcriptome via integrative analyses may provide more in-depth knowledge of the genomic landscape of CRC.
Collapse
Affiliation(s)
- Nur Zarina Ali Hassan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Norfilza Mohd Mokhtar
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- * E-mail: (NMM); (RJ)
| | - Teow Kok Sin
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Isa Mohamed Rose
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ismail Sagap
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Roslan Harun
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
- * E-mail: (NMM); (RJ)
| |
Collapse
|
18
|
Feiglin A, Ashkenazi S, Schlessinger A, Rost B, Ofran Y. Co-expression and co-localization of hub proteins and their partners are encoded in protein sequence. MOLECULAR BIOSYSTEMS 2014; 10:787-94. [PMID: 24457447 DOI: 10.1039/c3mb70411d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Spatiotemporal coordination is a critical factor in biological processes. Some hubs in protein-protein interaction networks tend to be co-expressed and co-localized with their partners more strongly than others, a difference which is arguably related to functional differences between the hubs. Based on numerous analyses of yeast hubs, it has been suggested that differences in co-expression and co-localization are reflected in the structural and molecular characteristics of the hubs. We hypothesized that if indeed differences in co-expression and co-localization are encoded in the molecular characteristics of the protein, it may be possible to predict the tendency for co-expression and co-localization of human hubs based on features learned from systematically characterized yeast hubs. Thus, we trained a prediction algorithm on hubs from yeast that were classified as either strongly or weakly co-expressed and co-localized with their partners, and applied the trained model to 800 human hub proteins. We found that the algorithm significantly distinguishes between human hubs that are co-expressed and co-localized with their partners and hubs that are not. The prediction is based on sequence derived features such as "stickiness", i.e. the existence of multiple putative binding sites that enable multiple simultaneous interactions, "plasticity", i.e. the existence of predicted structural disorder which conjecturally allows for multiple consecutive interactions with the same binding site and predicted subcellular localization. These results suggest that spatiotemporal dynamics is encoded, at least in part, in the amino acid sequence of the protein and that this encoding is similar in yeast and in human.
Collapse
Affiliation(s)
- Ariel Feiglin
- The Goodman faculty of life sciences, Bar Ilan University, Ramat Gan 52900, Israel.
| | | | | | | | | |
Collapse
|
19
|
Yu XY, Zhang Z, Zhang GJ, Guo KF, Kong CZ. Knockdown of Cdc25B in renal cell carcinoma is associated with decreased malignant features. Asian Pac J Cancer Prev 2012; 13:931-5. [PMID: 22631674 DOI: 10.7314/apjcp.2012.13.3.931] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Cdc25 phosphatases are important regulators of the cell cycle. Their abnormal expression detected in a number of tumors implies that their dysregulation is involved in malignant transformation. However, the role of Cdc25B in renal cell carcinomas remains unknown. To shed light on influence on renal cell carcinogenesis and subsequent progression, Cdc25B expression was examined by real-time RT-PCR and western blotting in renal cell carcinoma and normal tissues. 65 kDa Cdc25B expression was higher in carcinomas than in the adjacent normal tissues (P<0.05), positive correlations being noted with clinical stage and histopathologic grade (P<0.05). To additionally investigate the role of Cdc25B alteration in the development of renal cell carcinoma, Cdc25B siRNA was used to knockdown the expression of Cdc25B. Down-regulation resulted in slower growth, more G2/M cells, weaker capacity for migration and invasion, and induction of apoptosis in 769-P transfectants. Reduction of 14-3-3 protein expression appeared related to Cdc25B knockdown. These findings suggest an important role of Cdc25B in renal cell carcinoma development and provide a rationale for investigation of Cdc2B-based gene therapy.
Collapse
Affiliation(s)
- Xiu-Yue Yu
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | | | | | | | | |
Collapse
|
20
|
Li J, Zhang F, Chen JY. An integrated proteomics analysis of bone tissues in response to mechanical stimulation. BMC SYSTEMS BIOLOGY 2011; 5 Suppl 3:S7. [PMID: 22784626 PMCID: PMC3287575 DOI: 10.1186/1752-0509-5-s3-s7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Bone cells can sense physical forces and convert mechanical stimulation conditions into biochemical signals that lead to expression of mechanically sensitive genes and proteins. However, it is still poorly understood how genes and proteins in bone cells are orchestrated to respond to mechanical stimulations. In this research, we applied integrated proteomics, statistical, and network biology techniques to study proteome-level changes to bone tissue cells in response to two different conditions, normal loading and fatigue loading. We harvested ulna midshafts and isolated proteins from the control, loaded, and fatigue loaded Rats. Using a label-free liquid chromatography tandem mass spectrometry (LC-MS/MS) experimental proteomics technique, we derived a comprehensive list of 1,058 proteins that are differentially expressed among normal loading, fatigue loading, and controls. By carefully developing protein selection filters and statistical models, we were able to identify 42 proteins representing 21 Rat genes that were significantly associated with bone cells' response to quantitative changes between normal loading and fatigue loading conditions. We further applied network biology techniques by building a fatigue loading activated protein-protein interaction subnetwork involving 9 of the human-homolog counterpart of the 21 rat genes in a large connected network component. Our study shows that the combination of decreased anti-apoptotic factor, Raf1, and increased pro-apoptotic factor, PDCD8, results in significant increase in the number of apoptotic osteocytes following fatigue loading. We believe controlling osteoblast differentiation/proliferation and osteocyte apoptosis could be promising directions for developing future therapeutic solutions for related bone diseases.
Collapse
Affiliation(s)
- Jiliang Li
- Department of Biology, Purdue School of Science, Indiana University Purdue University Indianapolis (IUPUI), Indianapolis, IN 46202, USA
| | | | | |
Collapse
|
21
|
Jullien D, Bugler B, Dozier C, Cazales M, Ducommun B. Identification of N-terminally truncated stable nuclear isoforms of CDC25B that are specifically involved in G2/M checkpoint recovery. Cancer Res 2011; 71:1968-77. [PMID: 21363925 DOI: 10.1158/0008-5472.can-10-2453] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CDC25B phosphatases must activate cyclin B-CDK1 complexes to restart the cell cycle after an arrest in G2 phase caused by DNA damage. However, little is known about the precise mechanisms involved in this process, which may exert considerable impact on cancer susceptibility and therapeutic responses. Here we report the discovery of novel N-terminally truncated CDC25B isoforms, referred to as ΔN-CDC25B, with an exclusively nuclear and nonredundant function in cell cycle re-initiation after DNA damage. ΔN-CDC25B isoforms are expressed from a distinct promoter not involved in expression of canonical full-length isoforms. Remarkably, in contrast to the high lability and spatial dynamism of the full-length isoforms, ΔN-CDC25B isoforms are highly stable and exclusively nuclear, strongly suggesting the existence of two pools of CDC25B phosphatases in the cell that have functionally distinct properties. Using isoform-specific siRNA, we found that depleting full-length isoforms, but not ΔN-CDC25B isoforms, delays entry into mitosis. Thus, in an unperturbed cell cycle, the full-length isoforms are exclusively responsible for activating cyclin B-CDK1. Strikingly, in the late response to DNA damage, we found a CHK1-dependent shift in accumulation of CDC25B isoforms toward the ΔN-CDC25B species. Under this physiological stress condition, the ΔN-CDC25B isoform was found to play a crucial, nonredundant function in restarting the cell cycle after DNA damage-induced G2 phase arrest. Our findings reveal the existence of a previously unrecognized CDC25B isoform that operates specifically in the nucleus to reinitiate G2/M transition after DNA damage.
Collapse
|
22
|
Ji Y, Zhang YC, Pei LB, Shi LL, Yan JL, Ma XH. Anti-tumor effects of dihydroartemisinin on human osteosarcoma. Mol Cell Biochem 2011; 351:99-108. [PMID: 21234653 DOI: 10.1007/s11010-011-0716-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 01/04/2011] [Indexed: 11/25/2022]
Abstract
Dihydroartemisinin (DHA) exhibits antitumor activity against a wide spectrum of cancer cells. However, whether DHA has anti-tumor effect on human osteosarcoma cells remains unknown. This study aims to investigate the anti-tumor activity of DHA and the underlying mechanisms in human osteosarcoma cell lines with different p53 mutation statuses. Four human osteosarcoma cell lines were treated with different concentrations of DHA. Then, cell proliferation was determined by the CCK-8 viability assay; apoptosis and cell cycle progression were evaluated by flow cytometry; protein expression was analyzed by western blot assay; and NF-kB activity was examined by luciferase assay. The results demonstrated that DHA treatment could inhibit the proliferation of four osteosarcoma cell lines in a dose-dependent manner. P53 wild-type osteosarcoma cells were more sensitive to DHA. Moreover, the percentage of apoptotic cell and cell arrest in G₂/M phase was increased upon DHA treatment in a dose-dependent manner. Mechanistically, DHA activated caspase-3, caspase-8, and caspase-9; upregulated the expression of Bax, FAS, and cyclin D1; downregulated the expression of Bcl-2, Cdc25B, and cyclin B1; and inhibited the activity of NF-кB. In conclusion, DHA has significant anticancer effects against human osteosarcoma cells, which include induction of apoptosis and cell cycle arrest. The p53 gene may play a certain role in the DHA-induced human osteosarcoma apoptosis and cell cycle arrest. DHA is a novel anti-osteosarcoma drug candidate that merits further study.
Collapse
Affiliation(s)
- Ye Ji
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng St., Nangang District, Harbin, Heilongjiang 150001, China
| | | | | | | | | | | |
Collapse
|
23
|
Astuti P, Boutros R, Ducommun B, Gabrielli B. Mitotic phosphorylation of Cdc25B Ser321 disrupts 14-3-3 binding to the high affinity Ser323 site. J Biol Chem 2010; 285:34364-70. [PMID: 20801879 DOI: 10.1074/jbc.m110.138412] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cdc25B is a key regulator of entry into mitosis, and its activity and localization are regulated by binding of the 14-3-3 dimer. There are three 14-3-3 binding sites on Cdc25B, with Ser(323) being the highest affinity binding and is highly homologous to the Ser(216) 14-3-3 binding site on Cdc25C. Loss of 14-3-3 binding to Ser(323) increases cyclin/Cdk substrate access to the catalytic site, thereby increasing its activity. It also affects the localization of Cdc25B. Thus, phosphorylation and 14-3-3 binding to this site is essential for down-regulating Cdc25B activity, blocking its mitosis promoting function. The question of how this inhibitory signal is relieved to allow Cdc25B activation and entry into mitosis is yet to be resolved. Here, we show that Ser(323) phosphorylation is maintained into mitosis, but phosphorylation of Ser(321) disrupts 14-3-3 binding to Ser(323), mimicking the effect of inhibiting Ser(323) phosphorylation on both Cdc25B activity and localization. The unphosphorylated Ser(321) appears to have a role in stabilizing 14-3-3 binding to Ser(323), and loss of the Ser hydroxyl group appears to be sufficient to significantly reduce 14-3-3 binding. A consequence of loss of 14-3-3 binding is dephosphorylation of Ser(323). Ser(321) is phosphorylated in mitosis by Cdk1. The mitotic phosphorylation of Ser(321) acts to maintain full activation of Cdc25B by disrupting 14-3-3 binding to Ser(323) and enhancing the dephosphorylation of Ser(323) to block 14-3-3 binding to this site.
Collapse
Affiliation(s)
- Puji Astuti
- University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland 4102, Australia
| | | | | | | |
Collapse
|
24
|
Poehlmann A, Roessner A. Importance of DNA damage checkpoints in the pathogenesis of human cancers. Pathol Res Pract 2010; 206:591-601. [PMID: 20674189 DOI: 10.1016/j.prp.2010.06.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
All forms of life on earth must cope with constant exposure to DNA-damaging agents that may promote cancer development. As a biological barrier, known as DNA damage response (DDR), cells are provided with both DNA repair mechanisms and highly conserved cell cycle checkpoints. The latter are responsible for the control of cell cycle phase progression with ATM, ATR, Chk1, and Chk2 as the main signaling molecules, thus dealing with both endogenous and exogenous sources of DNA damage. As cell cycle checkpoint and also DNA repair genes, such as BRCA1 and BRCA2, are frequently mutated, we here discuss their fundamental roles in the pathogenesis of human cancers. Importantly, as current evidence also suggests a role of MAPK's (mitogen activated protein kinases) in cell cycle checkpoint control, we describe in this review both the ATR/ATM-Chk1/Chk2 signaling pathways as well as the regulation of cell cycle checkpoints by MAPK's as molecular mechanisms in DDR, and how their dysfunction is related to cancer development. Moreover, since damage to DNA might be the common underlying mechanism for the positive outcome of chemotherapy, we also discuss targeting anticancer treatments on cell cycle checkpoints as an important issue emerging in drug discovery.
Collapse
Affiliation(s)
- Angela Poehlmann
- Department of Pathology, Otto-von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | | |
Collapse
|
25
|
Lemaire M, Ducommun B, Nebreda AR. UV-induced downregulation of the CDC25B protein in human cells. FEBS Lett 2010; 584:1199-204. [DOI: 10.1016/j.febslet.2010.02.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 02/11/2010] [Accepted: 02/17/2010] [Indexed: 10/19/2022]
|
26
|
Bugler B, Schmitt E, Aressy B, Ducommun B. Unscheduled expression of CDC25B in S-phase leads to replicative stress and DNA damage. Mol Cancer 2010; 9:29. [PMID: 20128929 PMCID: PMC2825247 DOI: 10.1186/1476-4598-9-29] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 02/04/2010] [Indexed: 01/18/2023] Open
Abstract
Background CDC25B phosphatase is a cell cycle regulator that plays a critical role in checkpoint control. Up-regulation of CDC25B expression has been documented in a variety of human cancers, however, the relationships with the alteration of the molecular mechanisms that lead to oncogenesis still remain unclear. To address this issue we have investigated, in model cell lines, the consequences of unscheduled and elevated CDC25B levels. Results We report that increased CDC25B expression leads to DNA damage in the absence of genotoxic treatment. H2AX phosphorylation is detected in S-phase cells and requires active replication. We also report that CDC25B expression impairs DNA replication and results in an increased recruitment of the CDC45 replication factor onto chromatin. Finally, we observed chromosomal aberrations that are also enhanced upon CDC25B expression. Conclusion Overall, our results demonstrate that a moderate and unscheduled increase in CDC25B level, as observed in a number of human tumours, is sufficient to overcome the S-phase checkpoint efficiency thus leading to replicative stress and genomic instability.
Collapse
|
27
|
Lindqvist A, de Bruijn M, Macurek L, Brás A, Mensinga A, Bruinsma W, Voets O, Kranenburg O, Medema RH. Wip1 confers G2 checkpoint recovery competence by counteracting p53-dependent transcriptional repression. EMBO J 2009; 28:3196-206. [PMID: 19713933 PMCID: PMC2771084 DOI: 10.1038/emboj.2009.246] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Accepted: 07/29/2009] [Indexed: 01/07/2023] Open
Abstract
Activation of the DNA damage checkpoint causes a cell-cycle arrest through inhibition of cyclin-dependent kinases (cdks). To successfully recover from the arrest, a cell should somehow be maintained in its proper cell-cycle phase. This problem is particularly eminent when a cell arrests in G2, as cdk activity is important to establish a G2 state. Here, we identify the phosphatase Wip1 (PPM1D) as a factor that maintains a cell competent for cell-cycle re-entry during an ongoing DNA damage response in G2. We show that Wip1 function is required throughout the arrest, and that Wip1 acts by antagonizing p53-dependent repression of crucial mitotic inducers, such as Cyclin B and Plk1. Our data show that the primary function of Wip1 is to retain cellular competence to divide, rather than to silence the checkpoint to promote recovery. Our findings uncover Wip1 as a first in class recovery competence gene, and suggest that the principal function of Wip1 in cellular transformation is to retain proliferative capacity in the face of oncogene-induced stress.
Collapse
Affiliation(s)
- Arne Lindqvist
- Department of Medical Oncology and Cancer Genomics Center, Utrecht, Netherlands
| | - Menno de Bruijn
- Department of Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Libor Macurek
- Department of Medical Oncology and Cancer Genomics Center, Utrecht, Netherlands
| | - Alexandra Brás
- Genomic Instability Group, CNIO, C/Melchor Fernández Almagro, Madrid, Spain
| | - Anneloes Mensinga
- Department of Medical Oncology and Cancer Genomics Center, Utrecht, Netherlands
| | - Wytse Bruinsma
- Department of Medical Oncology and Cancer Genomics Center, Utrecht, Netherlands
| | - Olaf Voets
- Department of Medical Oncology and Cancer Genomics Center, Utrecht, Netherlands
| | - Onno Kranenburg
- Department of Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - René H Medema
- Department of Medical Oncology and Cancer Genomics Center, Utrecht, Netherlands
| |
Collapse
|
28
|
Uchida S, Yoshioka K, Kizu R, Nakagama H, Matsunaga T, Ishizaka Y, Poon RY, Yamashita K. Stress-Activated Mitogen-Activated Protein Kinases c-Jun NH2-Terminal Kinase and p38 Target Cdc25B for Degradation. Cancer Res 2009; 69:6438-44. [DOI: 10.1158/0008-5472.can-09-0869] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Lobjois V, Jullien D, Bouché JP, Ducommun B. The polo-like kinase 1 regulates CDC25B-dependent mitosis entry. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1793:462-8. [PMID: 19185590 DOI: 10.1016/j.bbamcr.2008.12.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 12/13/2008] [Accepted: 12/17/2008] [Indexed: 12/19/2022]
Abstract
Activation of cyclin-dependent kinase complexes (CDK) at key cell cycle transitions is dependent on their dephosphorylation by CDC25 dual-specificity phosphatases (CDC25A, B and C in human). The CDC25B phosphatase plays an essential role in controlling the activity of CDK1-cyclin B complexes at the entry into mitosis and together with polo-like kinase 1 (PLK1) in regulating the resumption of cell cycle progression after DNA damage-dependent checkpoint arrest in G2. In this study, we analysed the regulation of CDC25B-dependent mitosis entry by PLK1. We demonstrate that PLK1 activity is essential for the relocation of CDC25B from the cytoplasm to the nucleus. By gain and loss of function analyses, we show that PLK1 stimulates CDC25B-induced mitotic entry in both normal conditions and after DNA-damage induced G2/M arrest. Our results support a model in which the relocalisation of CDC25B to the nucleus at the G2-M transition by PLK1 regulates its mitotic inducing activity.
Collapse
Affiliation(s)
- Valerie Lobjois
- Université de Toulouse, LBCMCP, 118 Route de Narbonne, CNRS, LBCMCP-UMR5088, F-31062 Toulouse, France
| | | | | | | |
Collapse
|
30
|
Timofeev O, Cizmecioglu O, Hu E, Orlik T, Hoffmann I. Human Cdc25A phosphatase has a non-redundant function in G2 phase by activating Cyclin A-dependent kinases. FEBS Lett 2009; 583:841-7. [PMID: 19192479 DOI: 10.1016/j.febslet.2009.01.044] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Revised: 01/18/2009] [Accepted: 01/23/2009] [Indexed: 11/24/2022]
Abstract
Cdc25 phosphatases activate Cdk/Cyclin complexes by dephosphorylation and thus promote cell cycle progression. We observed that the peak activity of Cdc25A precedes the one of Cdc25B in prophase and the maximum of Cyclin/Cdk kinase activity. Furthermore, Cdc25A activates both Cdk1-2/Cyclin A and Cdk1/Cyclin B complexes while Cdc25B seems to be involved only in activation of Cdk1/Cyclin B. Concomitantly, repression of Cdc25A led to a decrease in Cyclin A-associated kinase activity and attenuated Cdk1 activation. Our results indicate that Cdc25A acts before Cdc25B - at least in cancer cells, and has non-redundant functions in late G2/early M-phase as a major regulator of Cyclin A/kinase complexes.
Collapse
Affiliation(s)
- Oleg Timofeev
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
31
|
Cui C, Zhao H, Zhang Z, Zong Z, Feng C, Zhang Y, Deng X, Xu X, Yu B. CDC25B acts as a potential target of PRKACA in fertilized mouse eggs. Biol Reprod 2008; 79:991-8. [PMID: 18633139 DOI: 10.1095/biolreprod.108.068205] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Protein kinase A (PRKACA) has been documented as a pivotal regulator in meiosis and mitosis arrest. Although our previous work has established that PRKACA regulates cell cycle progression of mouse fertilized eggs by inhibiting M-phase promoting factor (MPF), little is known about the intermediate factor between PRKACA and MPF in the mitotic cell cycle. In this study, we investigated the role of the PRKACA/CDC25B pathway on the early development of mouse fertilized eggs. Overexpression of unphosphorylatable CDC25B mutant (Cdc25b-S321A or Cdc25b-S229A/S321A) rapidly caused G2-phase eggs to enter mitosis. Microinjection of either Cdc25b-WT or Cdc25b-S229A mRNA also promoted G2/M transition, but much less efficiently than Cdc25b-S321A and Cdc25b-S229A/S321A. Moreover, mouse fertilized eggs overrode the G2 arrest by microinjection of either Cdc25b-S321A or Cdc25b-S229A/S321A mRNA, which efficiently resulted in MPF activation by directly dephosphorylating CDC2A-Tyr15, despite culture under conditions that maintained exogenous dibutyryl cAMP. Using a highly specific antibody against phospho-Ser321 of CDC25B in Western blotting, we showed that CDC25B-Ser321 was phosphorylated at the G1 and S phases, whereas Ser321 was dephosphorylated at the G2 and M phases in vivo. Our findings identify CDC25B as a potential target of PRKACA and show that PRKACA regulates G2/M transition by phosphorylating CDC25B-Ser321 but not CDC25B-Ser229 on the first mitotic division of mouse fertilized eggs.
Collapse
Affiliation(s)
- Cheng Cui
- Department of Physiology, China Medical University, Shenyang 110001, China
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Keaton MA, Szkotnicki L, Marquitz AR, Harrison J, Zyla TR, Lew DJ. Nucleocytoplasmic trafficking of G2/M regulators in yeast. Mol Biol Cell 2008; 19:4006-18. [PMID: 18562688 DOI: 10.1091/mbc.e08-03-0286] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Nucleocytoplasmic shuttling is prevalent among many cell cycle regulators controlling the G2/M transition. Shuttling of cyclin/cyclin-dependent kinase (CDK) complexes is thought to provide access to substrates stably located in either compartment. Because cyclin/CDK shuttles between cellular compartments, an upstream regulator that is fixed in one compartment could in principle affect the entire cyclin/CDK pool. Alternatively, the regulators themselves may need to shuttle to effectively regulate their moving target. Here, we identify localization motifs in the budding yeast Swe1p (Wee1) and Mih1p (Cdc25) cell cycle regulators. Replacement of endogenous Swe1p or Mih1p with mutants impaired in nuclear import or export revealed that the nuclear pools of Swe1p and Mih1p were more effective in CDK regulation than were the cytoplasmic pools. Nevertheless, shuttling of cyclin/CDK complexes was sufficiently rapid to coordinate nuclear and cytoplasmic events even when Swe1p or Mih1p were restricted to one compartment. Additionally, we found that Swe1p nuclear export was important for its degradation. Because Swe1p degradation is regulated by cytoskeletal stress, shuttling of Swe1p between nucleus and cytoplasm serves to couple cytoplasmic stress to nuclear cyclin/CDK inhibition.
Collapse
Affiliation(s)
- Mignon A Keaton
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
33
|
Sturgeon CM, Cinel B, Díaz-Marrero AR, McHardy LM, Ngo M, Andersen RJ, Roberge M. Abrogation of ionizing radiation-induced G2 checkpoint and inhibition of nuclear export by Cryptocarya pyrones. Cancer Chemother Pharmacol 2008; 61:407-13. [PMID: 17440726 DOI: 10.1007/s00280-007-0483-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Accepted: 03/17/2007] [Indexed: 10/23/2022]
Abstract
G(2) checkpoint inhibitors can force cells arrested in G(2) phase by DNA damage to enter mitosis. In this manner, several G(2) checkpoint inhibitors can enhance killing of cancer cells by ionizing radiation and DNA-damaging chemotherapeutic agents, particularly in cells lacking p53 function. All G(2) checkpoint inhibitors identified to date target protein phosphorylation by inhibiting checkpoint kinases or phosphatases. Using a phenotypic cell-based assay for G(2) checkpoint inhibitors, we have screened a large collection of plant extracts and identified Z-Cryptofolione and Cryptomoscatone D2 as highly efficacious inhibitors of the G(2) checkpoint. These compounds and related pyrones also inhibit nuclear export. Leptomycin B, a potent inhibitor of Crm1-mediated nuclear export, is also a very potent G(2) checkpoint inhibitor. These compounds possess a reactive Michael acceptor site and do not appear promising as a radiosensitizing agents because they are toxic to unirradiated cells at checkpoint inhibitory concentrations. Nevertheless, the results show that inhibition of nuclear export is an alternative to checkpoint kinase inhibition for abrogating the G(2) checkpoint and they should stimulate the search for less toxic nuclear export inhibitors.
Collapse
Affiliation(s)
- Christopher M Sturgeon
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | |
Collapse
|
34
|
Yan X, Chua MS, He J, So SK. Small interfering RNA targeting CDC25B inhibits liver tumor growth in vitro and in vivo. Mol Cancer 2008; 7:19. [PMID: 18269767 PMCID: PMC2276234 DOI: 10.1186/1476-4598-7-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Accepted: 02/12/2008] [Indexed: 01/09/2023] Open
Abstract
Background Using gene expression profiling, we previously identified CDC25B to be significantly highly expressed in hepatocellular carcinoma (HCC) compared to non-tumor liver. CDC25B is a cell cycle-activating phosphatase that positively regulates the activity of cyclin-dependent kinases, and is over-expressed in a variety of human malignancies. In this study, we validated the over-expression of CDC25B in HCC, and further investigated its potential as a therapeutic target for the management of HCC. Results Quantitative real-time polymerase chain reaction and immunohistochemical staining of patient samples confirmed the significant over-expression of CDC25B in HCC compared to non-tumor liver samples (P < 0.001). Thus, intefering with the expression and activity of CDC25B may be a potential way to intervene with HCC progression. We used RNA interference to study the biological effects of silencing CDC25B expression in HCC cell lines (Hep3B and Hep40), in order to validate its potential as a therapeutic target. Using small oligo siRNAs targeting the coding region of CDC25B, we effectively suppressed CDC25B expression by up to 90%. This was associatetd with significant reductions in cell growth rate, cell migration and invasion through the matrigel membrane, and caused significant cell cycle delay at the G2 phase. Finally, suppression of CDC25B significantly slowed the growth of Hep40 xenografts in nude mice. Conclusion Our data provide evidence that the inhibition of CDC25B expression and activity lead to suppression of tumor cell growth and motility, and may therefore be a feasible approach in the clinical management of HCC.
Collapse
Affiliation(s)
- Xinrui Yan
- Asian Liver Center, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
35
|
Didier C, Cavelier C, Quaranta M, Galcera MO, Demur C, Laurent G, Manenti S, Ducommun B. G2/M checkpoint stringency is a key parameter in the sensitivity of AML cells to genotoxic stress. Oncogene 2008; 27:3811-20. [PMID: 18212737 DOI: 10.1038/sj.onc.1211041] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute myeloid leukemia (AML) cells exposed to genotoxic agents arrest their cell cycle at the G2/M checkpoint and are inherently chemoresistant. To understand the mechanism of this chemoresistance, we compared the ability of immature CD34+ versus mature CD34- AML cell lines (KG1a and U937, respectively) to recover from a DNA damage-induced cell cycle checkpoint in G2. Here, we report that KG1a cells have a more stringent G2/M checkpoint response than U937 cells. We show that in both cell types, the CDC25B phosphatase participates in the G2/M checkpoint recovery and that its expression is upregulated. Furthermore, we show that CHK1 inhibition by UCN-01 in immature KG1a cells allows checkpoint exit and induces sensitivity to genotoxic agents. Similarly, UCN-01 treatment potentializes genotoxic-induced inhibition of colony formation efficiency of primary leukemic cells from AML patients. Altogether, our results demonstrate that checkpoint stringency varies during the maturation process and indicate that targeting checkpoint mechanisms might represent an attractive therapeutic opportunity for chemoresistant immature AML cells.
Collapse
Affiliation(s)
- C Didier
- LBCMCP-CNRS UMR5088-IFR109 Institut d'Exploration Fonctionnelle des Génomes, University of Toulouse, Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Lindqvist A, van Zon W, Karlsson Rosenthal C, Wolthuis RMF. Cyclin B1-Cdk1 activation continues after centrosome separation to control mitotic progression. PLoS Biol 2007; 5:e123. [PMID: 17472438 PMCID: PMC1858714 DOI: 10.1371/journal.pbio.0050123] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Accepted: 03/05/2007] [Indexed: 01/28/2023] Open
Abstract
Activation of cyclin B1–cyclin-dependent kinase 1 (Cdk1), triggered by a positive feedback loop at the end of G2, is the key event that initiates mitotic entry. In metaphase, anaphase-promoting complex/cyclosome–dependent destruction of cyclin B1 inactivates Cdk1 again, allowing mitotic exit and cell division. Several models describe Cdk1 activation kinetics in mitosis, but experimental data on how the activation proceeds in mitotic cells have largely been lacking. We use a novel approach to determine the temporal development of cyclin B1–Cdk1 activity in single cells. By quantifying both dephosphorylation of Cdk1 and phosphorylation of the Cdk1 target anaphase-promoting complex/cyclosome 3, we disclose how cyclin B1–Cdk1 continues to be activated after centrosome separation. Importantly, we discovered that cytoplasmic cyclin B1–Cdk1 activity can be maintained even when cyclin B1 translocates to the nucleus in prophase. These experimental data are fitted into a model describing cyclin B1–Cdk1 activation in human cells, revealing a striking resemblance to a bistable circuit. In line with the observed kinetics, cyclin B1–Cdk1 levels required to enter mitosis are lower than the amount of cyclin B1–Cdk1 needed for mitotic progression. We propose that gradually increasing cyclin B1–Cdk1 activity after centrosome separation is critical to coordinate mitotic progression. When active, the enzyme cyclin B1–cyclin-dependent kinase 1 (Cdk1) commits a growing cell to the process of mitotic cell division and chromosome separation. Cyclin B1–Cdk1 activation is controlled in many ways, but once its activity rises above a certain level, further activation of cyclin B1–Cdk1 is catalyzed by a positive-feedback loop. This generates highly active cyclin B1–Cdk1 and triggers the start of mitosis, which can only be completed when cyclin B1–Cdk1 activity is properly shut off again. However, it is not clear how cyclin B1–Cdk1 activity develops in human cells or how the switch between its inactive and active states is controlled. Our work combines activation measurements with a kinetic model to study how cyclin B1–Cdk1 activity accumulates just before and during mitosis. We show that cyclin B1–Cdk1 activity develops gradually in early mitosis and that different activity levels are required for initiation of, and progression through, mitosis. We also demonstrate that once cyclin B1–Cdk1 activation is truly launched, it is bound to continue and will not lightly drop back again. We propose that the successive cyclin B1–Cdk1 activity levels by themselves may coordinate the progression through the distinct phases of mitosis. The gradual increase of cyclin B1-Cdk1 activation in human cells is proposed to be critical for the progression of mitosis.
Collapse
Affiliation(s)
- Arne Lindqvist
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | |
Collapse
|
37
|
Bentley AM, Normand G, Hoyt J, King RW. Distinct sequence elements of cyclin B1 promote localization to chromatin, centrosomes, and kinetochores during mitosis. Mol Biol Cell 2007; 18:4847-58. [PMID: 17881737 PMCID: PMC2096604 DOI: 10.1091/mbc.e06-06-0539] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The mitotic cyclins promote cell division by binding and activating cyclin-dependent kinases (CDKs). Each cyclin has a unique pattern of subcellular localization that plays a vital role in regulating cell division. During mitosis, cyclin B1 is known to localize to centrosomes, microtubules, and chromatin. To determine the mechanisms of cyclin B1 localization in M phase, we imaged full-length and mutant versions of human cyclin B1-enhanced green fluorescent protein in live cells by using spinning disk confocal microscopy. In addition to centrosome, microtubule, and chromatin localization, we found that cyclin B1 also localizes to unattached kinetochores after nuclear envelope breakdown. Kinetochore recruitment of cyclin B1 required the kinetochore proteins Hec1 and Mad2, and it was stimulated by microtubule destabilization. Mutagenesis studies revealed that cyclin B1 is recruited to kinetochores through both CDK1-dependent and -independent mechanisms. In contrast, localization of cyclin B1 to chromatin and centrosomes is independent of CDK1 binding. The N-terminal domain of cyclin B1 is necessary and sufficient for chromatin association, whereas centrosome recruitment relies on sequences within the cyclin box. Our data support a role for cyclin B1 function at unattached kinetochores, and they demonstrate that separable and distinct sequence elements target cyclin B1 to kinetochores, chromatin, and centrosomes during mitosis.
Collapse
Affiliation(s)
- Anna M. Bentley
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Guillaume Normand
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Jonathan Hoyt
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Randall W. King
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
38
|
Feng C, Yu A, Liu Y, Zhang J, Zong Z, Su W, Zhang Z, Yu D, Sun QY, Yu B. Involvement of Protein Kinase B/AKT in Early Development of Mouse Fertilized Eggs1. Biol Reprod 2007; 77:560-8. [PMID: 17554083 DOI: 10.1095/biolreprod.107.060269] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The activation of AKT (also called protein kinase B) is thought to be a critical step in the phosphoinositide 3-kinase pathway that regulates cell growth and differentiation. In this report, we investigated the role of AKT in the regulation of mouse early embryo development. Injection of mRNA coding for a constitutively active myristoylated AKT (myr-Akt1) into one-cell stage fertilized eggs induced cell division more effectively than injection of wild-type AKT (Akt1-WT) mRNA, whereas microinjection of mRNA of kinase-deficient AKT (Akt1-KD) delayed the first mitotic division. Meanwhile, microinjection of different kinds of mRNA of AKT affected the phosphorylation status of CDC2A-Tyr15 and the activation of M-phase promoting factor (MPF). To investigate the intermediate factor between AKT and MPF, we then injected one-cell stage eggs first with Akt1-WT mRNA or myr-Akt1 mRNA and then with mRNA encoding either wild-type CDC25B (Cdc25b-WT) or a AKT-nonphosphorylatable Ser351 to Ala CDC25B mutant (Cdc25b-S351A). Cdc25b-S351A strongly inhibited the effect of AKT. Therefore, AKT causes the activation of MPF and strongly promotes the development of one-cell stage mouse fertilized eggs by inducing AKT-dependent phosphorylation of CDC25B, a member of the CDC25 phosphatase family. Our finding that CDC25B acts as a potential target of AKT provides new insight into the effect of AKT in the regulation of early development of mouse embryos.
Collapse
Affiliation(s)
- Chen Feng
- Department of Biochemical and Molecular Biology, China Medical University, Shenyang 110001, Liaoning, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Varmeh-Ziaie S, Manfredi JJ. The dual specificity phosphatase Cdc25B, but not the closely related Cdc25C, is capable of inhibiting cellular proliferation in a manner dependent upon its catalytic activity. J Biol Chem 2007; 282:24633-41. [PMID: 17591782 DOI: 10.1074/jbc.m703105200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cdc25B and Cdc25C are closely related dual specificity phosphatases that activate cyclin-dependent kinases by removal of inhibitory phosphorylations, thereby triggering entry into mitosis. Cdc25B, but not Cdc25C, has been implicated as an oncogene and been shown to be overexpressed in a variety of human tumors. Surprisingly, ectopic expression of Cdc25B, but not Cdc25C, inhibits cell proliferation in long term assays. Chimeric proteins generated from the two phosphatases show that the anti-proliferative activity is associated with the C-terminal end of Cdc25B. Indeed, the catalytic domain of Cdc25B is sufficient to suppress cell viability in a manner partially dependent upon its C-terminal 26 amino acids that is shown to influence substrate binding. Mutation analysis demonstrates that both the phosphatase activity of Cdc25B as well as its ability to interact with its substrates contribute to the inhibition of cell proliferation. These results demonstrate key differences in the biological activities of Cdc25B and Cdc25C caused by differential substrate affinity and recognition. This also argues that the antiproliferative activity of Cdc25B needs to be overcome for it to act as an oncogene during tumorigenesis.
Collapse
Affiliation(s)
- Shohreh Varmeh-Ziaie
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | |
Collapse
|
40
|
Kieffer I, Lorenzo C, Dozier C, Schmitt E, Ducommun B. Differential mitotic degradation of the CDC25B phosphatase variants. Oncogene 2007; 26:7847-58. [PMID: 17599046 DOI: 10.1038/sj.onc.1210596] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
CDC25 phosphatases control cell-cycle progression by dephosphorylating and activating cyclin-dependent kinases. CDC25B, one of the three members of this family in human cells, is thought to regulate initial mitotic events. CDC25B is an unstable protein whose proteasomal degradation is proposed to be controlled by beta-TrCP. Here, we have investigated the regulation of CDC25B during mitosis, using time-lapse video microscopy. We found that CDC25B expression is high during early mitosis, and that its degradation occurs after the metaphase-anaphase transition and cyclin B1 destruction. We also show that CDC25B degradation after metaphase is dependent on the integrity of the KEN-box and RRKSE motifs that are located within the alternatively spliced B domain, and that the CDC25B2 splice variant, that lacks this domain, is stable during mitosis. Furthermore, we show that the N-terminal region of CDC25B, encompassing the B domain, undergoes major conformational changes during mitosis that can be monitored by intramolecular fluorescence resonance energy transfer variation of specific CDC25B biosensors. This study demonstrates that CDC25B splice variants have differential mitotic stabilities, a feature that is likely to have major consequences on the local control of cyclin-dependent kinase-cyclin activities during mitotic progression.
Collapse
Affiliation(s)
- I Kieffer
- LBCMCP-CNRS UMR5088-IFR109 Institut d'Exploration Fonctionnelle des Génomes, Université Paul Sabatier, 118 route de Narbonne, Toulouse, France
| | | | | | | | | |
Collapse
|
41
|
Cuenda A, Rousseau S. p38 MAP-kinases pathway regulation, function and role in human diseases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1358-75. [PMID: 17481747 DOI: 10.1016/j.bbamcr.2007.03.010] [Citation(s) in RCA: 997] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 03/13/2007] [Accepted: 03/19/2007] [Indexed: 11/28/2022]
Abstract
Mammalian p38 mitogen-activated protein kinases (MAPKs) are activated by a wide range of cellular stresses as well as in response to inflammatory cytokines. There are four members of the p38MAPK family (p38alpha, p38beta, p38gamma and p38delta) which are about 60% identical in their amino acid sequence but differ in their expression patterns, substrate specificities and sensitivities to chemical inhibitors such as SB203580. A large body of evidences indicates that p38MAPK activity is critical for normal immune and inflammatory response. The p38MAPK pathway is a key regulator of pro-inflammatory cytokines biosynthesis at the transcriptional and translational levels, which makes different components of this pathway potential targets for the treatment of autoimmune and inflammatory diseases. However, recent studies have shed light on the broad effect of p38MAPK activation in the control of many other aspects of the physiology of the cell, such as control of cell cycle or cytoskeleton remodelling. Here we focus on these emergent roles of p38MAPKs and their implication in different pathologies.
Collapse
Affiliation(s)
- Ana Cuenda
- MRC Protein Phosphorylation Unit, College of life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK.
| | | |
Collapse
|
42
|
Jinlian L, Yingbin Z, Chunbo W. p38 MAPK in regulating cellular responses to ultraviolet radiation. J Biomed Sci 2007; 14:303-12. [PMID: 17334833 DOI: 10.1007/s11373-007-9148-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Accepted: 12/26/2006] [Indexed: 11/28/2022] Open
Abstract
Solar ultraviolet (UV) radiation is a major environmental factor that causes DNA damage, inflammation, erythema, sunburn, immunosuppression, photoaging, gene mutations, and skin cancer. p38 mitogen activated protein kinase (MAPK) are strongly activated by UV radiation, and play important roles in regulating cellular responses to UV. In this review, we examine the role played by p38 MAPK in mediating UV-induced cell cycle, apoptosis, inflammation, and skin tanning response. We review the role played by p38 MAPK in transcriptional regulation of key downstream genes that have been implicated in the regulation of cellular responses to UV radiation. Understanding this will undoubtedly help in the prevention and control of UV-induced damage and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Li Jinlian
- Medical College, Qingdao University, 422 Room, Boya Building, 308 Ningxia Road, Qingdao, 266071, China.
| | | | | |
Collapse
|
43
|
Schmidt A, Durgan J, Magalhaes A, Hall A. Rho GTPases regulate PRK2/PKN2 to control entry into mitosis and exit from cytokinesis. EMBO J 2007; 26:1624-36. [PMID: 17332740 PMCID: PMC1829391 DOI: 10.1038/sj.emboj.7601637] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Accepted: 02/05/2007] [Indexed: 01/20/2023] Open
Abstract
Rho GTPases regulate multiple signal transduction pathways that influence many aspects of cell behaviour, including migration, morphology, polarity and cell cycle. Through their ability to control the assembly and organization of the actin and microtubule cytoskeletons, Rho and Cdc42 make several key contributions during the mitotic phase of the cell cycle, including spindle assembly, spindle positioning, cleavage furrow contraction and abscission. We now report that PRK2/PKN2, a Ser/Thr kinase and Rho/Rac effector protein, is an essential regulator of both entry into mitosis and exit from cytokinesis in HeLa S3 cells. PRK2 is required for abscission of the midbody at the end of the cell division cycle and for phosphorylation and activation of Cdc25B, the phosphatase required for activation of mitotic cyclin/Cdk1 complexes at the G2/M transition. This reveals an additional step in the mammalian cell cycle controlled by Rho GTPases.
Collapse
Affiliation(s)
- Anja Schmidt
- Medical Research Council Laboratory for Molecular Cell Biology, Cancer Research UK Oncogene and Signal Transduction Group, University College London, London, UK
| | - Joanne Durgan
- Medical Research Council Laboratory for Molecular Cell Biology, Cancer Research UK Oncogene and Signal Transduction Group, University College London, London, UK
- Present address: Memorial Sloan-Kettering Cancer Center, 1275 York Avenue Box 572, New York, NY 10021, USA
| | - Ana Magalhaes
- Medical Research Council Laboratory for Molecular Cell Biology, Cancer Research UK Oncogene and Signal Transduction Group, University College London, London, UK
- Present address: Memorial Sloan-Kettering Cancer Center, 1275 York Avenue Box 572, New York, NY 10021, USA
| | - Alan Hall
- Medical Research Council Laboratory for Molecular Cell Biology, Cancer Research UK Oncogene and Signal Transduction Group, University College London, London, UK
- Present address: Memorial Sloan-Kettering Cancer Center, 1275 York Avenue Box 572, New York, NY 10021, USA
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue Box 572, New York, NY 10021, USA. Tel./Fax: +1 212 639 2387; E-mail:
| |
Collapse
|
44
|
Schmitt E, Boutros R, Froment C, Monsarrat B, Ducommun B, Dozier C. CHK1 phosphorylates CDC25B during the cell cycle in the absence of DNA damage. J Cell Sci 2006; 119:4269-75. [PMID: 17003105 DOI: 10.1242/jcs.03200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CDC25B is one of the three human phosphatases that activate the CDK-cyclin complexes, thereby triggering cell-cycle progression and division. Commitment to early mitotic events depends on the activation of a centrosomal pool of CDK1-cyclin-B1, and CDC25B is thought to be involved in initiating this centrosomal CDK1-cyclin-B1 activity. Centrosome-associated checkpoint kinase 1 (CHK1) has been proposed to contribute to the proper timing of a normal cell division cycle by inhibiting the activation of the centrosomal pool of CDK1. Here, we show that CDC25B is phosphorylated by CHK1 in vitro on multiple residues, including S230 and S563. We demonstrate these phosphorylations occur in vivo and that they are dependent on CHK1 activity. S230 CHK1-mediated phosphorylation is detected in cell extracts during S phase and G2 phase in the absence of DNA damage. We show that the S230-phosphorylated form of CDC25B is located at the centrosome from early S phase until mitosis. Furthermore, mutation of S230 to alanine increases the mitotic-inducing activity of CDC25B. Our results support a model in which, under normal cell cycle conditions and in the absence of DNA damage, CHK1 constitutively phosphorylates CDC25B during interphase and thus prevents the premature initiation of mitosis by negatively regulating the activity of CDC25B at the centrosome.
Collapse
Affiliation(s)
- Estelle Schmitt
- LBCMCP-CNRS UMR5088, IFR109, Université Paul Sabatier, 118 route de Narbonne, 31062 Toulouse, France
| | | | | | | | | | | |
Collapse
|
45
|
Karlsson-Rosenthal C, Millar JBA. Cdc25: mechanisms of checkpoint inhibition and recovery. Trends Cell Biol 2006; 16:285-92. [PMID: 16682204 DOI: 10.1016/j.tcb.2006.04.002] [Citation(s) in RCA: 196] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Accepted: 04/07/2006] [Indexed: 12/01/2022]
Abstract
Members of the eukaryotic Cdc25 phosphatase family are key targets of the Chk1 and Chk2 checkpoint kinases, which inactivate Cdc25 to halt cell cycle progression when DNA is damaged or incompletely replicated. Now, new kinases that phosphorylate and inactivate Cdc25 are being discovered, including MAPKAP kinase-2, a component of the p38 stress-activated MAP kinase pathway. The roles of other kinases, such as cyclin-dependent kinase, Polo and Aurora A kinase, in controlling the localization or the activation of Cdc25, are controversial. Here, we discuss new data that suggests that different Cdc25 isoforms and regulators of Cdc25 are differentially required for normal cell cycle progression and recovery from checkpoint arrest.
Collapse
|
46
|
Boutros R, Dozier C, Ducommun B. The when and wheres of CDC25 phosphatases. Curr Opin Cell Biol 2006; 18:185-91. [PMID: 16488126 DOI: 10.1016/j.ceb.2006.02.003] [Citation(s) in RCA: 294] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2005] [Accepted: 02/06/2006] [Indexed: 02/08/2023]
Abstract
The CDC25 phosphatases are key regulators of normal cell division and the cell's response to DNA damage. Earlier studies suggested non-overlapping roles for each isoform during a specific cell cycle phase. However, recent data suggest that multiple CDC25 isoforms cooperate to regulate each cell cycle transition. For instance, although CDC25A was initially thought to exclusively regulate the G(1)-S transition, recent data demonstrate a significant role for CDC25A in the G(2)-M transition. Further evidence demonstrates that in addition to the ATM/ATR-CHK pathway, a p38-MAPKAP pathway is also involved in controlling CDC25 activity during G(2)/M checkpoint activation. Together with the fact that CDC25 overexpression is reported in many cancers, these data highlight the significance of developing specific CDC25 inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Rose Boutros
- LBCMCP-CNRS UMR5088, IFR109, Université Paul Sabatier, 118 route de Narbonne, 31062 Toulouse, France
| | | | | |
Collapse
|
47
|
Lindqvist A, Källström H, Lundgren A, Barsoum E, Rosenthal CK. Cdc25B cooperates with Cdc25A to induce mitosis but has a unique role in activating cyclin B1-Cdk1 at the centrosome. ACTA ACUST UNITED AC 2005; 171:35-45. [PMID: 16216921 PMCID: PMC2171226 DOI: 10.1083/jcb.200503066] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cdc25 phosphatases are essential for the activation of mitotic cyclin–Cdks, but the precise roles of the three mammalian isoforms (A, B, and C) are unclear. Using RNA interference to reduce the expression of each Cdc25 isoform in HeLa and HEK293 cells, we observed that Cdc25A and -B are both needed for mitotic entry, whereas Cdc25C alone cannot induce mitosis. We found that the G2 delay caused by small interfering RNA to Cdc25A or -B was accompanied by reduced activities of both cyclin B1–Cdk1 and cyclin A–Cdk2 complexes and a delayed accumulation of cyclin B1 protein. Further, three-dimensional time-lapse microscopy and quantification of Cdk1 phosphorylation versus cyclin B1 levels in individual cells revealed that Cdc25A and -B exert specific functions in the initiation of mitosis: Cdc25A may play a role in chromatin condensation, whereas Cdc25B specifically activates cyclin B1–Cdk1 on centrosomes.
Collapse
Affiliation(s)
- Arne Lindqvist
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | | | | | | | | |
Collapse
|