101
|
Sinha D, Kalimutho M, Bowles J, Chan AL, Merriner DJ, Bain AL, Simmons JL, Freire R, Lopez JA, Hobbs RM, O'Bryan MK, Khanna KK. Cep55 overexpression causes male-specific sterility in mice by suppressing Foxo1 nuclear retention through sustained activation of PI3K/Akt signaling. FASEB J 2018; 32:4984-4999. [PMID: 29683733 DOI: 10.1096/fj.201701096rr] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Spermatogenesis is a dynamic process involving self-renewal and differentiation of spermatogonial stem cells, meiosis, and ultimately, the differentiation of haploid spermatids into sperm. Centrosomal protein 55 kDa (CEP55) is necessary for somatic cell abscission during cytokinesis. It facilitates equal segregation of cytoplasmic contents between daughter cells by recruiting endosomal sorting complex required for transport machinery (ESCRT) at the midbody. In germ cells, CEP55, in partnership with testes expressed-14 (TEX14) protein, has also been shown to be an integral component of intercellular bridge before meiosis. Various in vitro studies have demonstrated a role for CEP55 in multiple cancers and other diseases. However, its oncogenic potential in vivo remains elusive. To investigate, we generated ubiquitously overexpressing Cep55 transgenic ( Cep55Tg/Tg) mice aiming to characterize its oncogenic role in cancer. Unexpectedly, we found that Cep55Tg/Tg male mice were sterile and had severe and progressive defects in spermatogenesis related to spermatogenic arrest and lack of spermatids in the testes. In this study, we characterized this male-specific phenotype and showed that excessively high levels of Cep55 results in hyperactivation of PI3K/protein kinase B (Akt) signaling in testis. In line with this finding, we observed increased phosphorylation of forkhead box protein O1 (FoxO1), and suppression of its nuclear retention, along with the relative enrichment of promyelocytic leukemia zinc finger (PLZF) -positive cells. Independently, we observed that Cep55 amplification favored upregulation of ret ( Ret) proto-oncogene and glial-derived neurotrophic factor family receptor α-1 ( Gfra1). Consistent with these data, we observed selective down-regulation of genes associated with germ cell differentiation in Cep55-overexpressing testes at postnatal day 10, including early growth response-4 ( Egr4) and spermatogenesis and oogenesis specific basic helix-loop-helix-1 ( Sohlh1). Thus, Cep55 amplification leads to a shift toward the initial maintenance of undifferentiated spermatogonia and ultimately results in progressive germ cell loss. Collectively, our findings demonstrate that Cep55 overexpression causes change in germ cell proportions and manifests as a Sertoli cell only tubule phenotype, similar to that seen in many azoospermic men.-Sinha, D., Kalimutho, M., Bowles, J., Chan, A.-L., Merriner, D. J., Bain, A. L., Simmons, J. L., Freire, R., Lopez, J. A., Hobbs, R. M., O'Bryan, M. K., Khanna, K. K. Cep55 overexpression causes male-specific sterility in mice by suppressing Foxo1 nuclear retention through sustained activation of PI3K/Akt signaling.
Collapse
Affiliation(s)
- Debottam Sinha
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Natural Sciences, Griffith University, Nathan, Queensland, Australia
| | - Murugan Kalimutho
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Natural Sciences, Griffith University, Nathan, Queensland, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Ai-Leen Chan
- Germline Stem Cell Laboratory, Australian Regenerative Medicine Institute and Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - D Jo Merriner
- Male Infertility and Germ Cell Biology Laboratory, the School of Biological Sciences, Monash University, Clayton, Victoria, Australia; and
| | - Amanda L Bain
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Jacinta L Simmons
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas, Tenerife, Spain
| | - J Alejandro Lopez
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Natural Sciences, Griffith University, Nathan, Queensland, Australia
| | - Robin M Hobbs
- Germline Stem Cell Laboratory, Australian Regenerative Medicine Institute and Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Moira K O'Bryan
- Male Infertility and Germ Cell Biology Laboratory, the School of Biological Sciences, Monash University, Clayton, Victoria, Australia; and
| | - Kum Kum Khanna
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia
| |
Collapse
|
102
|
Qi J, Liu G, Wang F. High levels of centrosomal protein 55 expression is associated with poor clinical prognosis in patients with cervical cancer. Oncol Lett 2018; 15:9347-9352. [PMID: 29805659 DOI: 10.3892/ol.2018.8448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 03/07/2018] [Indexed: 01/03/2023] Open
Abstract
Centrosomal protein 55 (CEP55) has been proposed to have a role in tumor development. However, the expression pattern and clinical relevance of CEP55 has, to the best of our knowledge, not yet been investigated in cervical cancer. The mRNA levels of CEP55 in cervical cancer tissues and paired adjacent non-cancerous tissues were examined by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The present study assessed the association between immunohistochemical staining of CEP55 and clinicopathological characteristics and survival rates of patients. Compared with the adjacent non-cancerous tissues, CEP55 expression was significantly increased in cervical tumor tissues, as demonstrated by the results of RT-qPCR. High expression of CEP55 was significantly associated with lymph node metastasis (P=0.008) and advanced tumor stage (P=0.010). Furthermore, CEP55 overexpression in cervical cancer specimens was significantly associated with poor 5-year overall and recurrence-free survival rates (P=0.021 and P=0.010, respectively). The results of multivariate Cox regression analysis revealed that CEP55 expression was a significant, independent predictor for the survival of patients with cervical cancer (hazard ratio=3.057; P=0.035). These data indicated that high CEP55 expression was associated with lymph node metastasis and was an independent predictive factor for an unfavorable prognosis in patients with cervical carcinoma.
Collapse
Affiliation(s)
- Jingyi Qi
- Department of Reproductive Medicine, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Gelin Liu
- Department of Reproductive Medicine, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Fang Wang
- Department of Reproductive Medicine, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471000, P.R. China
| |
Collapse
|
103
|
Yin Y, Cai J, Meng F, Sui C, Jiang Y. MiR-144 suppresses proliferation, invasion, and migration of breast cancer cells through inhibiting CEP55. Cancer Biol Ther 2018; 19:306-315. [PMID: 29561704 PMCID: PMC5902245 DOI: 10.1080/15384047.2017.1416934] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/08/2017] [Accepted: 12/10/2017] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The study aimed to investigate the molecular mechanism of miR-144 and CEP55 as well as the influence of their interaction on the cell proliferation, migration, invasion, cell cycle and cell apoptosis in breast cancer. METHODS In this study, The Cancer Genome Atlas (TCGA, https://tcga-data.nci.nih.gov/ ) database was used for microarray analysis. The expressions of miR-144 and CEP55 in 40 adjacent tissues and 36 tumor tissues were examined by western blot, qRT-PCR and immunohistochemistry. The target relationship between miR-144 and CEP55 was predicted and confirmed by TargetScan and luciferase reporter assay. The cell proliferation, cell cycle and cell apoptosis in different groups were detected by MTT and flow cytometry assays, while wound healing and transwell assays were used for the cell migration and invasion tests. The regulatory effects of miR-144 and CEP55 on breast tumor were verified through nude mouse model in vivo experiment. RESULTS MiR-144 was down-regulated in breast cancerous tissues and cells, whereas CEP55 expression was up-regulated in breast cancerous tissues. Moreover, there existed a target relationship between miR-144 and CEP55 and negative correlation on their expressions. MiR-144 could down-regulate CEP55 expression, thereby inhibiting proliferation, invasion, migration, retarding cell cycle and accelerating cell apoptosis. MiR-144 could inhibit cell progression through down-regulating CEP55 in vivo. CONCLUSION MiR-144 suppressed cell proliferation, migration, invasion and induced cell cycle arrest and cell apoptosis by repressing CEP55. This might provide a promising therapy for clinical treatment.
Collapse
Affiliation(s)
- Yuanqin Yin
- Biotherapy Laboratory, Cancer Institute, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingjing Cai
- Department of Medicine, General Hospital of Fushun Mining Bureau, Fushun, Liaoning, China
| | - Fandong Meng
- Biotherapy Laboratory, Cancer Institute, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chengguang Sui
- Biotherapy Laboratory, Cancer Institute, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Youhong Jiang
- Biotherapy Laboratory, Cancer Institute, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
104
|
Li F, Jin D, Tang C, Gao D. CEP55 promotes cell proliferation and inhibits apoptosis via the PI3K/Akt/p21 signaling pathway in human glioma U251 cells. Oncol Lett 2018; 15:4789-4796. [PMID: 29552118 PMCID: PMC5840555 DOI: 10.3892/ol.2018.7934] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022] Open
Abstract
Human glioma is one of the major malignancies worldwide with an increased mortality rate. Centrosomal protein of 55 kDa (CEP55) is an essential component of the CEP family and has been identified as a prognostic marker for multiple types of cancer. However, the function of CEP55 during glioma tumorigenesis remains unclear. In the present study, the data derived from the Oncomine database indicated that the expression of CEP55 is increased in glioma tissues compared with normal tissues. Furthermore, the expression of CEP55 was also increased at the level of mRNA and protein in glioma cell lines compared with normal human astrocytes. The knockdown of CEP55 expression inhibited the proliferation of glioma U251 cells, whereas overexpression of CEP55 induced the proliferation of U251 cells. Flow cytometric analysis indicated that the knockdown of CEP55 resulted in an increased number of cells arrested at G2/M phase, and apoptosis was promoted. Further investigations revealed that the overexpression of CEP55 increased the phosphorylation of Akt and inhibited the activity of p21. By contrast, the knockdown of CEP55 resulted in the opposite effects. Taken together, the results of the present study suggested that CEP55 regulated the proliferation of glioma cells, further attributing to the carcinogenesis and progression of glioma via the PI3K/Akt/p21 signaling pathway. Therefore, CEP55 may be a novel therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Feng Li
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Dan Jin
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Otolaryngology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Chuanxi Tang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Dianshuai Gao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
105
|
Maine EA, Westcott JM, Prechtl AM, Dang TT, Whitehurst AW, Pearson GW. The cancer-testis antigens SPANX-A/C/D and CTAG2 promote breast cancer invasion. Oncotarget 2018; 7:14708-26. [PMID: 26895102 PMCID: PMC4924746 DOI: 10.18632/oncotarget.7408] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/29/2016] [Indexed: 12/19/2022] Open
Abstract
Genes that are normally biased towards expression in the testis are often induced in tumor cells. These gametogenic genes, known as cancer-testis antigens (CTAs), have been extenstively investigated as targets for immunotherapy. However, despite their frequent detection, the degree to which CTAs support neoplastic invasion is poorly understood. Here, we find that the CTA genes SPANX-A/C/D and CTAG2 are coordinately induced in breast cancer cells and regulate distinct features of invasive behavior. Our functional analysis revealed that CTAG2 interacts with Pericentrin at the centrosome and is necessary for directional migration. Conversely, SPANX-A/C/D interacts with Lamin A/C at the inner nuclear membrane and is required for the formation of actin-rich cellular protrusions that reorganize the extracellular matrix. Importantly, SPANX-A/C/D was required for breast cancer cells to spontaneously metastasize to the lung, demonstrating that CTA reactivation can be critical for invasion dependent phenotypes in vivo. Moreover, elevated SPANX-A/C/D expression in breast cancer patient tumors correlated with poor outcome. Together, our results suggest that distinct CTAs promote tumor progression by regulating complementary cellular functions that are integrated together to induce invasive behavior.
Collapse
Affiliation(s)
- Erin A Maine
- Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jill M Westcott
- Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Amanda M Prechtl
- Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tuyen T Dang
- Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Angelique W Whitehurst
- Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,The Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Gray W Pearson
- Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,The Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
106
|
Zhu H, Chen D, Tang J, Huang C, Lv S, Wang D, Li G. Overexpression of centrosomal protein 55 regulates the proliferation of glioma cell and mediates proliferation promoted by EGFRvIII in glioblastoma U251 cells. Oncol Lett 2017; 15:2700-2706. [PMID: 29434995 DOI: 10.3892/ol.2017.7573] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 11/23/2017] [Indexed: 12/28/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is often amplified in glioma, with the most common extracellular domain mutation being EGFR variant III (EGFRvIII). Abnormal EGFRvIII signaling has been shown to be important in driving tumor progression. Centrosomal protein 55 (CEP55), a member of the centrosomal relative proteins family, participates cytokinesis in the cell cycle. It exists in a few normal tissues and various tumor cells. The expression and function of CEP55 in human glioma cells need to investigate. In this study, the expression of CEP55 was detected in 40 cases of glioma tissues and 10 cases of non-tumor brain tissue. The proliferation of glioblastoma U251 cells was analyzed after transfection with EGFRvIII and CEP55 siRNA. We found that the expression of CEP55 was increased significantly in the glioma tissues than in normal brain tissue. The proliferation of U251 cells increased remarkably after transfection with EGFRvIII. Knockdown of CEP55 inhibited proliferation of U251 cells and was able to eliminate the effect of promoting proliferation induced by EGFRvIII in U251 cells. CEP55 played a key role in the proliferation of glioma cells and mediated EGFRvIII-stimulated proliferation in glioma cells. CEP55 might be a novel molecular therapeutic target in patients with gliomas expressing EGFRvIII.
Collapse
Affiliation(s)
- Hongfan Zhu
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, The Third Military Medical University, P.R. China
| | - Diangang Chen
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, The Third Military Medical University, P.R. China
| | - Jinliang Tang
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, P.R. China
| | - Changlin Huang
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, The Third Military Medical University, P.R. China
| | - Shengqing Lv
- Department of Neurosurgery, Xinqiao Hospital, The Third Military Medical University, P.R. China
| | - Donglin Wang
- Department of Oncology, Cancer Hospital of Chongqing, Chongqing 400037, P.R. China
| | - Guanghui Li
- Institute for Cancer Research in People's Liberation Army, Xinqiao Hospital, The Third Military Medical University, P.R. China
| |
Collapse
|
107
|
Stoten CL, Carlton JG. ESCRT-dependent control of membrane remodelling during cell division. Semin Cell Dev Biol 2017; 74:50-65. [PMID: 28843980 PMCID: PMC6015221 DOI: 10.1016/j.semcdb.2017.08.035] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/07/2017] [Accepted: 08/18/2017] [Indexed: 12/16/2022]
Abstract
The Endosomal Sorting Complex Required for Transport (ESCRT) proteins form an evolutionarily conserved membrane remodelling machinery. Identified originally for their role in cargo sorting and remodelling of endosomal membranes during yeast vacuolar sorting, an extensive body of work now implicates a sub-complex of this machinery (ESCRT-III), as a transplantable membrane fission machinery that is dispatched to various cellular locations to achieve a topologically unique membrane separation. Surprisingly, several ESCRT-III-regulated processes occur during cell division, when cells undergo a dramatic and co-ordinated remodelling of their membranes to allow the physical processes of division to occur. The ESCRT machinery functions in regeneration of the nuclear envelope during open mitosis and in the abscission phase of cytokinesis, where daughter cells are separated from each other in the last act of division. Roles for the ESCRT machinery in cell division are conserved as far back as Archaea, suggesting that the ancestral role of these proteins was as a membrane remodelling machinery that facilitated division and that was co-opted throughout evolution to perform a variety of other cell biological functions. Here, we will explore the function and regulation of the ESCRT machinery in cell division.
Collapse
|
108
|
Growing functions of the ESCRT machinery in cell biology and viral replication. Biochem Soc Trans 2017; 45:613-634. [PMID: 28620025 DOI: 10.1042/bst20160479] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 01/31/2023]
Abstract
The vast expansion in recent years of the cellular processes promoted by the endosomal sorting complex required for transport (ESCRT) machinery has reinforced its identity as a modular system that uses multiple adaptors to recruit the core membrane remodelling activity at different intracellular sites and facilitate membrane scission. Functional connections to processes such as the aurora B-dependent abscission checkpoint also highlight the importance of the spatiotemporal regulation of the ESCRT machinery. Here, we summarise the role of ESCRTs in viral budding, and what we have learned about the ESCRT pathway from studying this process. These advances are discussed in the context of areas of cell biology that have been transformed by research in the ESCRT field, including cytokinetic abscission, nuclear envelope resealing and plasma membrane repair.
Collapse
|
109
|
Peng T, Zhou W, Guo F, Wu HS, Wang CY, Wang L, Yang ZY. Centrosomal protein 55 activates NF-κB signalling and promotes pancreatic cancer cells aggressiveness. Sci Rep 2017; 7:5925. [PMID: 28724890 PMCID: PMC5517556 DOI: 10.1038/s41598-017-06132-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/07/2017] [Indexed: 01/05/2023] Open
Abstract
Centrosomal protein 55 (CEP55) is a microtubule-bundling protein that participants in cell mitosis. It is overexpressed in several solid tumours and promotes the growth and invasion of cancer cells. However, the role of CEP55 in pancreatic cancer (PANC) remains unclear. Herein, upregulated expression of CEP55 (associated with poor prognosis) was detected in PANC using quantitative real-time reverse transcription PCR, western blotting, and immunohistochemistry. Cell migration, colony formation, wound-healing, and Transwell matrix penetration assays, revealed that upregulation of CEP55 promoted PANC cells proliferation, migration, and invasion in vitro, whereas knockdown of CEP55 attenuated it. In an in vivo murine model, CEP55 overexpression accelerated PANC cells tumourigenicity, together with upregulation of the protein levels of invasion-related proteins matrix metalloproteinase (MMP)2, MMP9, and proliferation-related protein Cyclin D1. Downregulation of CEP55 had the reverse effect. Moreover, the nuclear factor κB (NF-κB)/IκBα signalling pathway, which was activated in CEP55-transduced PANC cells and inhibited in CEP55-silenced PANC cells, contributed to CEP55-mediated PANC cell aggressiveness. This study provided new insights into the oncogenic roles of CEP55 and the mechanism by which the NF-κB pathway is hyperactivated in patients with PANC, indicating that CEP55 is a valuable prognostic factor and a potential therapeutic target in PANC.
Collapse
Affiliation(s)
- Tao Peng
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Wei Zhou
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Feng Guo
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - He-Shui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Chun-You Wang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Li Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Zhi-Yong Yang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
110
|
Wang Q, Tang Y, Xu Y, Xu S, Jiang Y, Dong Q, Zhou Y, Ge W. The X-linked deubiquitinase USP9X is an integral component of centrosome. J Biol Chem 2017; 292:12874-12884. [PMID: 28620049 DOI: 10.1074/jbc.m116.769943] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 06/02/2017] [Indexed: 12/23/2022] Open
Abstract
The X-linked deubiquitinase USP9X has been implicated in multiple pathological disorders including malignancies and X-linked intellectual disability. However, its biological function and substrate repertoire remain to be investigated. In this study, we utilized the tandem mass tag labeling assay to identify USP9X-regulated proteins and revealed that the expression of multiple genes is altered in USP9X-deficient cells. Interestingly, we showed that USP9X promotes stabilization of centrosome proteins PCM1 and CEP55 through its catalytic activity. Remarkably, we demonstrated that USP9X is physically associated and spatially co-localized with PCM1 and CEP55 in the centrosome, and we revealed that either PCM1 or CEP55 loss resulted in impairment of USP9X centrosome localization. Moreover, we showed that USP9X is required for centrosome duplication, and this effect is dependent on its catalytic activity and its N-terminal module, which is responsible for physical association of USP9X with PCM1 and CEP55. Collectively, our experiments identified USP9X as an integral component of the centrosome where it functions to stabilize PCM1 and CEP55 and promote centrosome biogenesis.
Collapse
Affiliation(s)
- Qian Wang
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yiman Tang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yue Xu
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shilei Xu
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yong Jiang
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Qiuping Dong
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China.
| | - Wenshu Ge
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| |
Collapse
|
111
|
Kumar S, Sharma G, Chakraborty C, Sharma AR, Kim J. Regulatory functional territory of PLK-1 and their substrates beyond mitosis. Oncotarget 2017; 8:37942-37962. [PMID: 28415805 PMCID: PMC5514964 DOI: 10.18632/oncotarget.16290] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/03/2017] [Indexed: 12/04/2022] Open
Abstract
Polo-like kinase 1 (PLK-1) is a well-known (Ser/Thr) mitotic protein kinase and is considered as a proto-oncogene. As hyper-activation of PLK-1 is broadly associated with poor prognosis and cancer progression, it is one of the most extensively studied mitotic kinases. During mitosis, PLK-1 regulates various cell cycle events, such as spindle pole maturation, chromosome segregation and cytokinesis. However, studies have demonstrated that the role of PLK-1 is not only restricted to mitosis, but PLK-1 can also regulate other vital events beyond mitosis, including transcription, translation, ciliogenesis, checkpoint adaptation and recovery, apoptosis, chromosomes dynamics etc. Recent reviews have tried to define the regulatory role of PLK-1 during mitosis progression and tumorigenesis, but its' functional role beyond mitosis is still largely unexplored. PLK-1 can regulate the activity of many proteins that work outside of its conventional territory. The dysregulation of these proteins can cause diseases such as Alzheimer's disease, tumorigenesis etc. and may also lead to drug resistance. Thus, in this review, we discussed the versatile role of PLK-1 and tried to collect data to validate its' functional role in cell cycle regulation apart from mitosis.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Garima Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Chiranjib Chakraborty
- Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Ashish Ranjan Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| |
Collapse
|
112
|
Ott CM. Midbody remnant licenses primary cilia formation in epithelial cells. J Cell Biol 2017; 214:237-9. [PMID: 27482049 PMCID: PMC4970334 DOI: 10.1083/jcb.201607046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 07/13/2016] [Indexed: 11/22/2022] Open
Abstract
Tethered midbody remnants dancing across apical microvilli, encountering the centrosome, and beckoning forth a cilium-who would have guessed this is how polarized epithelial cells coordinate the end of mitosis and the beginning of ciliogenesis? New evidence from Bernabé-Rubio et al. (2016. J. Cell Biol http://dx.doi.org/10.1083/jcb.201601020) supports this emerging model.
Collapse
Affiliation(s)
- Carolyn M Ott
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| |
Collapse
|
113
|
A nonsense mutation inCEP55defines a new locus for a Meckel-like syndrome, an autosomal recessive lethal fetal ciliopathy. Clin Genet 2017; 92:510-516. [DOI: 10.1111/cge.13012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/09/2017] [Accepted: 03/03/2017] [Indexed: 01/05/2023]
|
114
|
Thieleke-Matos C, Osório DS, Carvalho AX, Morais-de-Sá E. Emerging Mechanisms and Roles for Asymmetric Cytokinesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 332:297-345. [PMID: 28526136 DOI: 10.1016/bs.ircmb.2017.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cytokinesis completes cell division by physically separating the contents of the mother cell between the two daughter cells. This event requires the highly coordinated reorganization of the cytoskeleton within a precise window of time to ensure faithful genomic segregation. In addition, recent progress in the field highlighted the importance of cytokinesis in providing particularly important cues in the context of multicellular tissues. The organization of the cytokinetic machinery and the asymmetric localization or inheritance of the midbody remnants is critical to define the spatial distribution of mechanical and biochemical signals. After a brief overview of the conserved steps of animal cytokinesis, we review the mechanisms controlling polarized cytokinesis focusing on the challenges of epithelial cytokinesis. Finally, we discuss the significance of these asymmetries in defining embryonic body axes, determining cell fate, and ensuring the correct propagation of epithelial organization during proliferation.
Collapse
Affiliation(s)
- C Thieleke-Matos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cell Division and Genomic stability, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - D S Osório
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cytoskeletal Dynamics, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A X Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cytoskeletal Dynamics, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - E Morais-de-Sá
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cell Division and Genomic stability, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
115
|
Frosk P, Arts HH, Philippe J, Gunn CS, Brown EL, Chodirker B, Simard L, Majewski J, Fahiminiya S, Russell C, Liu YP, Hegele R, Katsanis N, Goerz C, Del Bigio MR, Davis EE. A truncating mutation in CEP55 is the likely cause of MARCH, a novel syndrome affecting neuronal mitosis. J Med Genet 2017; 54:490-501. [PMID: 28264986 PMCID: PMC5502313 DOI: 10.1136/jmedgenet-2016-104296] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/08/2016] [Accepted: 12/14/2016] [Indexed: 11/17/2022]
Abstract
Background Hydranencephaly is a congenital anomaly leading to replacement of the cerebral hemispheres with a fluid-filled cyst. The goals of this work are to describe a novel autosomal-recessive syndrome that includes hydranencephaly (multinucleated neurons, anhydramnios, renal dysplasia, cerebellar hypoplasia and hydranencephaly (MARCH)); to identify its genetic cause(s) and to provide functional insight into pathomechanism. Methods We used homozygosity mapping and exome sequencing to identify recessive mutations in a single family with three affected fetuses. Immunohistochemistry, RT-PCR and imaging in cell lines, and zebrafish models, were used to explore the function of the gene and the effect of the mutation. Results We identified a homozygous nonsense mutation in CEP55 segregating with MARCH. Testing the effect of this allele on patient-derived cells indicated both a reduction of the overall CEP55 message and the production of a message that likely gives rise to a truncated protein. Suppression or ablation of cep55l in zebrafish embryos recapitulated key features of MARCH, most notably renal dysplasia, cerebellar hypoplasia and craniofacial abnormalities. These phenotypes could be rescued by full-length but not truncated human CEP55 message. Finally, we expressed the truncated form of CEP55 in human cells, where we observed a failure of truncated protein to localise to the midbody, leading to abscission failure and multinucleated daughter cells. Conclusions CEP55 loss of function mutations likely underlie MARCH, a novel multiple congenital anomaly syndrome. This association expands the involvement of centrosomal proteins in human genetic disorders by highlighting a role in midbody function.
Collapse
Affiliation(s)
- Patrick Frosk
- Departments of Pediatrics and Child Health, University of Manitoba, Manitoba, Canada.,Departments of Biochemistry and Medical Genetics, University of Manitoba, Manitoba, Canada
| | - Heleen H Arts
- Departments of Biochemistry, University of Western Ontario, London, Ontario, Canada.,Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Julien Philippe
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | - Carter S Gunn
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | - Emma L Brown
- Departments of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Bernard Chodirker
- Departments of Pediatrics and Child Health, University of Manitoba, Manitoba, Canada.,Departments of Biochemistry and Medical Genetics, University of Manitoba, Manitoba, Canada
| | - Louise Simard
- Departments of Biochemistry and Medical Genetics, University of Manitoba, Manitoba, Canada
| | - Jacek Majewski
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Somayyeh Fahiminiya
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Chad Russell
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | - Yangfan P Liu
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Robert Hegele
- Departments of Biochemistry, University of Western Ontario, London, Ontario, Canada.,Departments of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | - Conrad Goerz
- Departments of Pathology, University of Manitoba, Manitoba, Canada
| | - Marc R Del Bigio
- Departments of Pathology, University of Manitoba, Manitoba, Canada.,Diagnostic Services Manitoba, Manitoba, Canada
| | - Erica E Davis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
116
|
Jiang W, Wang Z, Jia Y. CEP55 overexpression predicts poor prognosis in patients with locally advanced esophageal squamous cell carcinoma. Oncol Lett 2016; 13:236-242. [PMID: 28123547 PMCID: PMC5244839 DOI: 10.3892/ol.2016.5414] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/27/2016] [Indexed: 12/29/2022] Open
Abstract
Development of esophageal squamous cell carcinoma (ESCC) involves alterations in multiple genes with corresponding proteins. Recent studies have demonstrated that centrosomal protein 55 (CEP55) shares certain features with oncogenes, and CEP55 overexpression is associated with the development and progression of malignant tumors. The present study aimed to analyze, for the first time, whether CEP55 expression is related to clinicopothalogic features in the esophageal squamous cell carcinoma (ESCC), as well as patient survival. A total of 110 patients with mid-thoracic ESCC who suffered from Ivor-Lewis were enrolled. The CEP55 expression profile of these patients in tumour tissues and corresponding healthy esophageal mucosa (CHEM) was detected by immunohistochemistry and semi-quantitative reverse transcription-polymerase chain reaction analyses. Correlations between CEP55 expression and clinicopathological factors were analyzed using χ2 test. The log-rank test was employed to calculate survival rate. A Cox regression multivariate analysis was performed to determine independent prognostic factors. The results demonstrated that CEP55 expression in ESCC was significantly higher than that of CHEM (P<0.001). Overexpression of CEP55 was significantly associated with differentiation degree (P=0.022), T stage (P=0.019), lymph node metastasis (P=0.033), clinicopathological staging (P=0.002) and tumor recurrence (P=0.021) in locally advanced ESCC patients. In addition, CEP55 overexpression was significantly associated with reduced overall survival of patients after surgery (P=0.012). The 5-year survival rate of patients without CEP55 overexpression was significantly higher than that of patients with CEP55 overexpression (P=0.012). Therefore, these findings suggest that CEP55 overexpression correlates with poor prognosis in locally advanced ESCC patients.
Collapse
Affiliation(s)
- Wenpeng Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Zhou Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yang Jia
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
117
|
Arnold N, Girke T, Sureshchandra S, Nguyen C, Rais M, Messaoudi I. Genomic and functional analysis of the host response to acute simian varicella infection in the lung. Sci Rep 2016; 6:34164. [PMID: 27677639 PMCID: PMC5039758 DOI: 10.1038/srep34164] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/08/2016] [Indexed: 01/19/2023] Open
Abstract
Varicella Zoster Virus (VZV) is the causative agent of varicella and herpes zoster. Although it is well established that VZV is transmitted via the respiratory route, the host-pathogen interactions during acute VZV infection in the lungs remain poorly understood due to limited access to clinical samples. To address these gaps in our knowledge, we leveraged a nonhuman primate model of VZV infection where rhesus macaques are intrabronchially challenged with the closely related Simian Varicella Virus (SVV). Acute infection is characterized by immune infiltration of the lung airways, a significant up-regulation of genes involved in antiviral-immunity, and a down-regulation of genes involved in lung development. This is followed by a decrease in viral loads and increased expression of genes associated with cell cycle and tissue repair. These data provide the first characterization of the host response required to control varicella virus replication in the lung and provide insight into mechanisms by which VZV infection can cause lung injury in an immune competent host.
Collapse
Affiliation(s)
- Nicole Arnold
- Graduate Program in Microbiology, University of California-Riverside, CA, USA
| | - Thomas Girke
- Department of Botany and Plant Sciences, University of California-Riverside, CA, USA
| | - Suhas Sureshchandra
- Graduate Program in Genetics, Genomics and Bioinformatics, University of California-Riverside, CA, USA
| | - Christina Nguyen
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA, USA
| | - Ilhem Messaoudi
- Graduate Program in Microbiology, University of California-Riverside, CA, USA
- Graduate Program in Genetics, Genomics and Bioinformatics, University of California-Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA, USA
| |
Collapse
|
118
|
Liu L, Mei Q, Zhao J, Dai Y, Fu Q. Suppression of CEP55 reduces cell viability and induces apoptosis in human lung cancer. Oncol Rep 2016; 36:1939-1945. [PMID: 27633074 DOI: 10.3892/or.2016.5059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/12/2016] [Indexed: 11/05/2022] Open
Abstract
Centrosomal protein 55 (CEP55), identified as a centrosome‑associated protein, has been reported to be involved in human malignancies. However, its biological function in human lung cancer remains largely unknown. In the present study, we firstly analyzed the expression of CEP55 in 20 pairs of lung cancer and matched non‑tumor tissues using quantitative RT‑PCR analysis and found that CEP55 mRNA was significantly increased in lung cancer tissues compared with that in matched tumor‑adjacent tissues. Then we performed a loss‑of‑function assay using lung cancer cell lines A549 and 95D. Functionally, knockdown of CEP55 markedly inhibited cell viability and proliferation ability as determined by MTT and colony formation assays. Moreover, CEP55‑silenced cells were obviously arrested in the G0/G1 phase and presented significant cell apoptosis as determined using flow cytometric analysis. Mechanistically, western blot analysis further revealed that knockdown of CEP55 decreased the expression of CDK4, p21 and Bcl‑2, while it increased the expression of pro‑apoptotic protein, Bad, caspase‑3 and PARP in 95D cells. In conclusion, our data highlight the crucial role of CEP55 in promoting lung cancer cell proliferation in vitro and its inhibition may be a novel therapeutic strategy for lung cancer.
Collapse
Affiliation(s)
- Ligang Liu
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qi Mei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jing Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuhong Dai
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qiang Fu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
119
|
Li X, Tran KM, Aziz KE, Sorokin AV, Chen J, Wang W. Defining the Protein-Protein Interaction Network of the Human Protein Tyrosine Phosphatase Family. Mol Cell Proteomics 2016; 15:3030-44. [PMID: 27432908 DOI: 10.1074/mcp.m116.060277] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Indexed: 12/25/2022] Open
Abstract
Protein tyrosine phosphorylation, which plays a vital role in a variety of human cellular processes, is coordinated by protein tyrosine kinases and protein tyrosine phosphatases (PTPs). Genomic studies provide compelling evidence that PTPs are frequently mutated in various human cancers, suggesting that they have important roles in tumor suppression. However, the cellular functions and regulatory machineries of most PTPs are still largely unknown. To gain a comprehensive understanding of the protein-protein interaction network of the human PTP family, we performed a global proteomic study. Using a Minkowski distance-based unified scoring environment (MUSE) for the data analysis, we identified 940 high confidence candidate-interacting proteins that comprise the interaction landscape of the human PTP family. Through a gene ontology analysis and functional validations, we connected the PTP family with several key signaling pathways or cellular functions whose associations were previously unclear, such as the RAS-RAF-MEK pathway, the Hippo-YAP pathway, and cytokinesis. Our study provides the first glimpse of a protein interaction network for the human PTP family, linking it to a number of crucial signaling events, and generating a useful resource for future studies of PTPs.
Collapse
Affiliation(s)
- Xu Li
- From the ‡Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030
| | - Kim My Tran
- From the ‡Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030
| | - Kathryn E Aziz
- From the ‡Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030
| | - Alexey V Sorokin
- From the ‡Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030
| | - Junjie Chen
- From the ‡Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030;
| | - Wenqi Wang
- §Department of Developmental and Cell Biology, University of California, Irvine, California 92697
| |
Collapse
|
120
|
Wang G, Liu M, Wang H, Yu S, Jiang Z, Sun J, Han K, Shen J, Zhu M, Lin Z, Jiang C, Guo M. Centrosomal Protein of 55 Regulates Glucose Metabolism, Proliferation and Apoptosis of Glioma Cells via the Akt/mTOR Signaling Pathway. J Cancer 2016; 7:1431-1440. [PMID: 27471559 PMCID: PMC4964127 DOI: 10.7150/jca.15497] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/18/2016] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Glioma is one of the most common and most aggressive brain tumors in humans. The molecular and cellular mechanisms responsible for the onset and the progression of glioma are elusive and controversial. Centrosomal protein of 55 (CEP55) was initially described as a highly coiled-coil protein that plays critical roles in cell division, but was recently identified as being overexpressed in many human cancers. The function of CEP55 has not previously been characterized in glioma. We aim to discover the effect and mechanism of CEP55 in glioma development. METHOD qRT-PCR and immunohistochemistry were used to analyze CEP55 expression. Glucose uptake, western blot, MTS, CCK-8, Caspase-3 activity and TUNEL staining assays were performed to investigate the role and mechanism of CEP55 on glioma cell process. RESULTS We found that the levels of CEP55 expression were upregulated in glioma. In addition, CEP55 appeared to regulate glucose metabolism of glioma cells. Furthermore, knockdown of CEP55 inhibited cell proliferation and induced cell apoptosis in glioma. Finally, we provided preliminary evidence that knockdown of CEP55 inhibited glioma development via suppressing the activity of Akt/mTOR signaling. CONCLUSIONS Our results demonstrated that CEP55 regulates glucose metabolism, proliferation and apoptosis of glioma cells via the Akt/mTOR signaling pathway, and its promotive effect on glioma tumorigenesis can be a potential target for glioma therapy in the future.
Collapse
Affiliation(s)
- Guangzhi Wang
- 1. Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
- 2. Department of Medical Service Management, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Mingna Liu
- 3. Department of Gastroenterology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Hongjun Wang
- 1. Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Shan Yu
- 4. Department of Pathology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Zhenfeng Jiang
- 5. Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Jiahang Sun
- 1. Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Ke Han
- 6. School of Computer and Information Engineering, Harbin University of Commerce, Harbin, Heilongjiang 150086, China
| | - Jia Shen
- 7. Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Minwei Zhu
- 5. Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Zhiguo Lin
- 5. Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Chuanlu Jiang
- 1. Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Mian Guo
- 1. Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| |
Collapse
|
121
|
Kamranvar SA, Gupta DK, Huang Y, Gupta RK, Johansson S. Integrin signaling via FAK-Src controls cytokinetic abscission by decelerating PLK1 degradation and subsequent recruitment of CEP55 at the midbody. Oncotarget 2016; 7:30820-30. [PMID: 27127172 PMCID: PMC5058720 DOI: 10.18632/oncotarget.9003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 04/09/2016] [Indexed: 01/08/2023] Open
Abstract
Adhesion to extracellular matrix is required for cell cycle progression through the G1 phase and for the completion of cytokinesis in normal adherent cells. Cancer cells acquire the ability to proliferate anchorage-independently, a characteristic feature of malignantly transformed cells. However, the molecular mechanisms underlying this escape of the normal control mechanisms remain unclear. The current study aimed to identify adhesion-induced reactions regulating the cytokinesis of non-transformed human fibroblasts.The adhesion-dependent control of cytokinesis was found to occur at a late stage close to the abscission, during which the endosomal sorting complex required for transport (ESCRT) severs the thin intercellular bridge connecting two nascent daughter cells. CEP55, a key protein involved in the abscission process, was localized at the midbody in both adherent and non-adherent fibroblasts, but it was unable to efficiently recruit ALIX, TSG101, and consequently the ESCRT-III subunit CHMP4B was missing in the non-adherent cells. PLK1, a kinase that prevents premature recruitment of CEP55 to the midbody, disappeared from this site more rapidly in the non-adherent cells. A FAK-Src signaling pathway downstream of integrin-mediated cell adhesion was found to decelerate both PLK1 degradation and CEP55 accumulation at the midbody. These data identify the regulation of PLK1 and CEP55 as steps where integrins exert control over the cytokinetic abscission.
Collapse
Affiliation(s)
- Siamak A. Kamranvar
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Deepesh Kumar Gupta
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Ying Huang
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Rajesh Kumar Gupta
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Staffan Johansson
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
122
|
Panchenko MV. Structure, function and regulation of jade family PHD finger 1 (JADE1). Gene 2016; 589:1-11. [PMID: 27155521 DOI: 10.1016/j.gene.2016.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/28/2016] [Accepted: 05/01/2016] [Indexed: 12/24/2022]
Abstract
The family of JADE proteins includes three paralogues encoded by individual genes and designated PHF17 (JADE1), PHF16 (JADE2), and PHF15 (JADE3). All three JADE proteins bear in tandem two Plant Homeo-domains (PHD) which are zinc finger domains. This review focuses on one member of the JADE family, JADE1. Studies addressing the biochemical, cellular and biological role of JADE1 are discussed. Recent discoveries of JADE1 function in the regulation of the epithelial cell cycle with potential relevance to disease are presented. Unresolved questions and future directions are formulated.
Collapse
Affiliation(s)
- Maria V Panchenko
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, United States.
| |
Collapse
|
123
|
Kumar S, Sharma AR, Sharma G, Chakraborty C, Kim J. PLK-1: Angel or devil for cell cycle progression. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1865:190-203. [PMID: 26899266 DOI: 10.1016/j.bbcan.2016.02.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/01/2016] [Accepted: 02/16/2016] [Indexed: 12/31/2022]
Abstract
PLK-1 is a key player in the eukaryotic cell cycle. Cell cycle progression is precisely controlled by cell cycle regulatory kinases. PLK-1 is a mitotic kinase that actively regulates the G2/M transition, mitosis, mitotic exit, and cytokinesis. During cell cycle progression, PLK-1 controls various events related to the cell cycle maturation, directly and/or indirectly. On the contrary, aberrant expression of PLK-1 is strongly associated with tumorigenesis and its poor prognosis. The misexpression of PLK-1 causes the abnormalities including aneuploidy, mitotic defects, leading to tumorigenesis through inhibiting the p53 and pRB genes. Therefore, we reviewed the role of PLK-1 in the cell cycle progression and in the tumorigenesis either as a cell cycle regulator or on an attractive anti-cancer drug target.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo 200-704, Republic of Korea.
| | - Ashish Ranjan Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo 200-704, Republic of Korea.
| | - Garima Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo 200-704, Republic of Korea.
| | - Chiranjib Chakraborty
- Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida 203201, India.
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo 200-704, Republic of Korea.
| |
Collapse
|
124
|
Okamoto A, Yabuta N, Mukai S, Torigata K, Nojima H. Phosphorylation of CHO1 by Lats1/2 regulates the centrosomal activation of LIMK1 during cytokinesis. Cell Cycle 2016; 14:1568-82. [PMID: 25786116 DOI: 10.1080/15384101.2015.1026489] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Large tumor suppressor 1 and 2 (Lats1/2) regulate centrosomal integrity, chromosome segregation and cytokinesis. As components of the centralspindlin complex, the kinesin-like protein CHO1 and its splicing variant MKLP1 colocalize with chromosome passenger proteins and GTPases and regulate the formation of the contractile ring and cytokinesis; however, the regulatory mechanisms of CHO1/MKLP1 remain elusive. Here, we show that Lats1/2 phosphorylate Ser716 in the F-actin-interacting region of CHO1, which is absent in MKLP1. Phosphorylated CHO1 localized to the centrosomes and midbody, and the actin polymerization factor LIM-kinase 1 (LIMK1) was identified as its binding partner. Overexpression of constitutively phosphorylated and non-phosphorylated CHO1 altered the mitotic localization and activation of LIMK1 at the centrosomes in HeLa cells, leading to the inhibition of cytokinesis through excessive phosphorylation of Cofilin and mislocalization of Ect2. These results suggest that Lats1/2 stringently control cytokinesis by regulating CHO1 phosphorylation and the mitotic activation of LIMK1 on centrosomes.
Collapse
Affiliation(s)
- Ayumi Okamoto
- a Department of Molecular Genetics ; Research Institute for Microbial Diseases; Osaka University ; Suita City , Osaka , Japan
| | | | | | | | | |
Collapse
|
125
|
Wang Y, Jin T, Dai X, Xu J. Lentivirus-mediated knockdown of CEP55 suppresses cell proliferation of breast cancer cells. Biosci Trends 2016; 10:67-73. [PMID: 26902787 DOI: 10.5582/bst.2016.01010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Centrosomal protein 55 (CEP55), as a microtubule-bundling protein, plays an important role in cell cycle regulation. CEP55 has been recognized recently in several human cancers. In this study, we first observed that the mRNA level of CEP55 is commonly up-regulated in breast cancer compared with their normal counterparts as demonstrated by data derived from Oncomine database. To further evaluate the functional role of CEP55 in breast cancer cells. Expression of CEP55 was efficiently knocked down using lentivirus-mediated RNA interference in human breast cancer cell line ZR-75-30, as evidenced by quantitative real-time PCR (qRT-PCR) and Western blot analysis. Further investigations revealed that CEP55 knockdown significantly inhibited cell proliferation and colony formation. Moreover, flow cytometer analysis indicated knockdown of CEP55 induced cell cycle arrested at G0/G1 phase and cell apoptosis. These findings suggest that CEP55 plays a crucial role in promoting breast cancer cell proliferation and it might be a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Ye Wang
- Department of general surgery, Shanghai first people's Hospital, Shanghai Jiao Tong University School of Medicine
| | | | | | | |
Collapse
|
126
|
Ratsima H, Serrano D, Pascariu M, D'Amours D. Centrosome-Dependent Bypass of the DNA Damage Checkpoint by the Polo Kinase Cdc5. Cell Rep 2016; 14:1422-1434. [PMID: 26832404 DOI: 10.1016/j.celrep.2016.01.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/01/2015] [Accepted: 12/30/2015] [Indexed: 11/15/2022] Open
Abstract
Cell-cycle checkpoints are essential feedback mechanisms that promote genome integrity. However, in the face of unrepairable DNA lesions, bypass mechanisms can suppress checkpoint activity and allow cells to resume proliferation. The molecular mechanisms underlying this biological response are currently not understood. Taking advantage of unique separation-of-function mutants, we show that the Polo-like kinase (PLK) Cdc5 uses a phosphopriming-based interaction mechanism to suppress G2/M checkpoint arrest by targeting Polo kinase activity to centrosomes. We also show that key subunits of the evolutionarily conserved RSC complex are critical downstream effectors of Cdc5 activity in checkpoint suppression. Importantly, the lethality and checkpoint defects associated with loss of Cdc5 Polo box activity can be fully rescued by artificially anchoring Cdc5 kinase domain to yeast centrosomes. Collectively, our results highlight a previously unappreciated role for centrosomes as key signaling centers for the suppression of cell-cycle arrest induced by persistent or unrepairable DNA damage.
Collapse
Affiliation(s)
- Hery Ratsima
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada; Département de Pathologie et Biologie Cellulaire, Université de Montréal, P.O. Box 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Diego Serrano
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada; Département de Pathologie et Biologie Cellulaire, Université de Montréal, P.O. Box 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Mirela Pascariu
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Damien D'Amours
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada; Département de Pathologie et Biologie Cellulaire, Université de Montréal, P.O. Box 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada.
| |
Collapse
|
127
|
Jiang W, Wang Z, Chen G, Jia Y. Prognostic significance of centrosomal protein 55 in stage I pulmonary adenocarcinoma after radical resection. Thorac Cancer 2016; 7:316-22. [PMID: 27148417 PMCID: PMC4846620 DOI: 10.1111/1759-7714.12330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 11/26/2015] [Indexed: 11/29/2022] Open
Abstract
Background Pulmonary adenocarcinoma is a predominant pathologic non‐small cell lung cancer (NSCLC) with a high morbidity in China. Even at histological stage I, many patients still experience recurrence after radical surgery; therefore, it is critical to determine useful indicators to stratify patients according to recurrent risk. Centrosomal protein 55 (CEP55) shares certain characteristics with oncogenes and aberrant expression of CEP55 can lead to tumorigenesis. Therefore, we aimed to clarify the clinicopathological significance and prognostic value of CEP55 in stage I pulmonary adenocarcinoma. Methods We enrolled 106 patients with stage I pulmonary adenocarcinoma who had received complete resection in our study. CEP55 expression levels in the pulmonary tissues of all patients were validated by Western blot analyses and immunohistochemistry. SPSS 17.0 software was employed to analyze the correlation between CEP55 expression and clinicopathological characteristics of patients, as well as prognosis. Results CEP55 overexpression was detected in 67 patients (63.2%). Overexpression is associated with tumor differentiation (P = 0.036), T stage (P = 0.000) and visceral pleural invasion (P = 0.009). Patients with CEP55 overexpression had worse survival compared with those with low expression (P = 0.043). Univariate analysis revealed that T stage (P = 0.000), differentiation degree (P = 0.002), visceral pleural invasion (P = 0.000), and tumor size (P = 0.013) were also significant prognostic factors. Conclusion CEP55 is a useful predicator to improve stratification of patients with stage I pulmonary adenocarcinoma.
Collapse
Affiliation(s)
- Wenpeng Jiang
- Department of Thoracic Surgery Shandong Provincial Hospital affiliated to Shandong University Jinan Shandong China
| | - Zhou Wang
- Department of Thoracic Surgery Shandong Provincial Hospital affiliated to Shandong University Jinan Shandong China
| | - Gang Chen
- Department of Thoracic Surgery Shandong Provincial Hospital affiliated to Shandong University Jinan Shandong China
| | - Yang Jia
- Department of Thoracic Surgery Shandong Provincial Hospital affiliated to Shandong University Jinan Shandong China
| |
Collapse
|
128
|
Fischer M, Quaas M, Nickel A, Engeland K. Indirect p53-dependent transcriptional repression of Survivin, CDC25C, and PLK1 genes requires the cyclin-dependent kinase inhibitor p21/CDKN1A and CDE/CHR promoter sites binding the DREAM complex. Oncotarget 2015; 6:41402-17. [PMID: 26595675 PMCID: PMC4747163 DOI: 10.18632/oncotarget.6356] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 11/11/2015] [Indexed: 12/15/2022] Open
Abstract
The transcription factor p53 is central to cell cycle control by downregulation of cell cycle-promoting genes upon cell stress such as DNA damage. Survivin (BIRC5), CDC25C, and PLK1 encode important cell cycle regulators that are repressed following p53 activation. Here, we provide evidence that p53-dependent repression of these genes requires activation of p21 (CDKN1A, WAF1, CIP1). Chromatin immunoprecipitation (ChIP) data indicate that promoter binding of B-MYB switches to binding of E2F4 and p130 resulting in a replacement of the MMB (Myb-MuvB) by the DREAM complex. We demonstrate that this replacement depends on p21. Furthermore, transcriptional repression by p53 requires intact DREAM binding sites in the target promoters. The CDE and CHR cell cycle promoter elements are the sites for DREAM binding. These elements as well as the p53 response of Survivin, CDC25C, and PLK1 are evolutionarily conserved. No binding of p53 to these genes is detected by ChIP and mutation of proposed p53 binding sites does not alter the p53 response. Thus, a mechanism for direct p53-dependent transcriptional repression is not supported by the data. In contrast, repression by DREAM is consistent with most previous findings and unifies models based on p21-, E2F4-, p130-, and CDE/CHR-dependent repression by p53. In conclusion, the presented data suggest that the p53-p21-DREAM-CDE/CHR pathway regulates p53-dependent repression of Survivin, CDC25C, and PLK1.
Collapse
Affiliation(s)
- Martin Fischer
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
- Department of Medical Oncology, Dana–Farber Cancer Institute, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marianne Quaas
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Annina Nickel
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Kurt Engeland
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| |
Collapse
|
129
|
Xu ZY, Ma XS, Qi ST, Wang ZB, Guo L, Schatten H, Sun QY, Sun YP. Cep55 regulates spindle organization and cell cycle progression in meiotic oocyte. Sci Rep 2015; 5:16978. [PMID: 26582107 PMCID: PMC4652202 DOI: 10.1038/srep16978] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/22/2015] [Indexed: 12/29/2022] Open
Abstract
Cep55 is a relatively novel member of the centrosomal protein family. Here, we show that Cep55 is expressed in mouse oocytes from the germinal vesicle (GV) to metaphase II (MII) stages. Immuostaining and confocal microscopy as well as time lapse live imaging after injection of mRNA encoding fusion protein of Cep55 and GFP identified that Cep55 was localized to the meiotic spindle, especially to the spindle poles at metaphase, while it was concentrated at the midbody in telophase in meiotic oocytes. Knockdown of Cep55 by specific siRNA injection caused the dissociation of γ-tubulin from the spindle poles, resulting in severely defective spindles and misaligned chromosomes, leading to metaphase I arrest and failure of first polar body (PB1) extrusion. Correspondingly, cyclin B accumulation and spindle assembly checkpoint (SAC) activation were observed in Cep55 knockdown oocytes. Our results suggest that Cep55 may act as an MTOC-associated protein regulating spindle organization, and thus cell cycle progression during mouse oocyte meiotic maturation.
Collapse
Affiliation(s)
- Zhao-Yang Xu
- The Reproductive Medical Center, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052 China
| | - Xue-Shan Ma
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020 China
| | - Shu-Tao Qi
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020 China
| | - Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020 China
| | - Lei Guo
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020 China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri-Columbia, MO 65211, USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020 China
| | - Ying-Pu Sun
- The Reproductive Medical Center, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052 China
| |
Collapse
|
130
|
Hui L, Yang N, Yang H, Guo X, Jang X. Identification of biomarkers with a tumor stage-dependent expression and exploration of the mechanism involved in laryngeal squamous cell carcinoma. Oncol Rep 2015; 34:2627-35. [PMID: 26323359 DOI: 10.3892/or.2015.4230] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/02/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to identify biomarkers with a tumor stage-dependent expression manner and explore the regulatory mechanisms of laryngeal squamous cell carcinoma (LSCC) progression. Microarray data GSE59102 was used for differential analysis using a limma package. Enrichment analyses were performed for the differentially expressed genes (DEGs) between tumor tissues and normal tissues at different stages. A co-expressed network involving the overlapped DEGs in two stages was established based on Pearson's correlation coefficients. Furthermore, for the tumor stage‑dependent expressed DEGs, a protein‑protein interaction (PPI) network was constructed by mapping the genes using the STRING database. Transcription factors (TFs), oncogenes and tumor‑associated genes (TSGs) among the DEGs were predicted, following a search of the TRANSFAC, tumor-associated gene (TAG) and TSG databases. The CDT database was used to identify LSCC‑associated genes. In total, 696 DEGs from early stage and control samples and 622 DEGs from advanced sttage and control samples were selected, which were mainly enriched in the cell cycle pathway. In the co-expressed network, BUB1, TTK, E2F1 and CEP55 were prominent, with E2F1 being predicted as a TSG and CEP55 as an oncogene. The HOX family members were predicted as TFs. MMP1, MMP9, MMP3 and PLAU were the most evident nodes in the PPI network, where MMP3 was connected with MMP1. The ADH family was correlated with LSCC. Several biomarkers with tumor stage-dependent expression were identified including MMP1, MMP3, MMP9, PLAU and ADHs. Additionally, the dysregulated cell cycle pathway involving BUB1, TTK, E2F1 and CEP55, and the mediation of MMP1 by MMP3 as well as the predicted TF HOX, may all play significant roles in LSCC progression.
Collapse
Affiliation(s)
- Lian Hui
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ning Yang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Huijun Yang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xing Guo
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xuejun Jang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
131
|
Siriwardana NS, Meyer RD, Panchenko MV. The novel function of JADE1S in cytokinesis of epithelial cells. Cell Cycle 2015; 14:2821-34. [PMID: 26151225 DOI: 10.1080/15384101.2015.1068476] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
JADE1 belongs to a small family of PHD zinc finger proteins that interacts with histone acetyl transferase (HAT) HBO1 and is associated with chromatin. We recently reported JADE1 chromatin shuttling and phosphorylation during G2/M to G1 transition, which was sensitive to Aurora A inhibition. In the current study we examined mechanisms of the cell cycle regulation by the small isoform of JADE1 protein, JADE1S, and report data showing that JADE1S has a novel function in the regulation of cytokinesis. Using FACS assays, we show that, JADE1S depletion facilitated rates of G1-cells accumulation in synchronously dividing HeLa cell cultures. Depletion of JADE1S protein in asynchronously dividing cells decreased the proportion of cytokinetic cells, and increased the proportion of multi-nuclear cells, indicative of premature and failed cytokinesis. In contrast, moderate overexpression of JADE1S increased the number of cytokinetic cells in time- and dose- dependent manner, indicating cytokinetic delay. Pharmacological inhibition of Aurora B kinase resulted in the release of JADE1S-mediated cytokinetic delay and allowed progression of abscission in cells over-expressing JADE1S. Finally, we show that JADE1S protein localized to centrosomes in interphase and mitotic cells, while during cytokinesis JADE1S localized to the midbody. Neither JADE1L nor partner of JADE1, HAT HBO1 was localized to the centrosomes or midbodies. Our study identifies the novel role for JADE1S in regulation of cytokinesis and suggests function in Aurora B kinase-mediated cytokinesis checkpoint.
Collapse
|
132
|
Jeffery J, Neyt C, Moore W, Paterson S, Bower NI, Chenevix‐Trench G, Verkade H, Hogan BM, Khanna KK. Cep55 regulates embryonic growth and development by promoting Akt stability in zebrafish. FASEB J 2015; 29:1999-2009. [DOI: 10.1096/fj.14-265090] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Jessie Jeffery
- Signal Transduction Laboratory, QIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Christine Neyt
- Vascular Biology and Development Laboratory, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQueenslandAustralia
| | - Wade Moore
- Zebrafish Developmental Genetics Laboratory, Monash UniversityClaytonVictoriaAustralia
| | - Scott Paterson
- Vascular Biology and Development Laboratory, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQueenslandAustralia
| | - Neil I. Bower
- Vascular Biology and Development Laboratory, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQueenslandAustralia
| | - Georgia Chenevix‐Trench
- Cancer Genetics Laboratory, QIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Heather Verkade
- Zebrafish Developmental Genetics Laboratory, Monash UniversityClaytonVictoriaAustralia
| | - Benjamin M. Hogan
- Vascular Biology and Development Laboratory, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQueenslandAustralia
| | - Kum Kum Khanna
- Signal Transduction Laboratory, QIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| |
Collapse
|
133
|
Jeffery J, Sinha D, Srihari S, Kalimutho M, Khanna KK. Beyond cytokinesis: the emerging roles of CEP55 in tumorigenesis. Oncogene 2015; 35:683-90. [PMID: 25915844 DOI: 10.1038/onc.2015.128] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/16/2015] [Accepted: 03/16/2015] [Indexed: 01/10/2023]
Abstract
CEP55 was initially identified as a pivotal component of abscission, the final stage of cytokinesis, serving to regulate the physical separation of two daughter cells. Over the past 10 years, several studies have illuminated additional roles for CEP55 including regulating the PI3K/AKT pathway and midbody fate. Concurrently, CEP55 has been studied in the context of cancers including those of the breast, lung, colon and liver. CEP55 overexpression has been found to significantly correlate with tumor stage, aggressiveness, metastasis and poor prognosis across multiple tumor types and therefore has been included as part of several prognostic 'gene signatures' for cancer. Here by discussing in depth the functions of CEP55 across different effector pathways, and also its roles as a biomarker and driver of tumorigenesis, we assemble an exhaustive review, thus commemorating a decade of research on CEP55.
Collapse
Affiliation(s)
- J Jeffery
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - D Sinha
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Natural Sciences, Griffith University, Brisbane, Queensland, Australia
| | - S Srihari
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - M Kalimutho
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - K K Khanna
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
134
|
St-Denis N, Gupta GD, Lin ZY, Gonzalez-Badillo B, Pelletier L, Gingras AC. Myotubularin-related proteins 3 and 4 interact with polo-like kinase 1 and centrosomal protein of 55 kDa to ensure proper abscission. Mol Cell Proteomics 2015; 14:946-60. [PMID: 25659891 PMCID: PMC4390272 DOI: 10.1074/mcp.m114.046086] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
The myotubularins are a family of phosphatases that dephosphorylate the phosphatidylinositols phosphatidylinositol-3-phosphate and phosphatidylinositol-3,5-phosphate. Several family members are mutated in disease, yet the biological functions of the majority of myotubularins remain unknown. To gain insight into the roles of the individual enzymes, we have used affinity purification coupled to mass spectrometry to identify protein-protein interactions for the myotubularins. The myotubularin interactome comprises 66 high confidence (false discovery rate ≤1%) interactions, including 18 pairwise interactions between individual myotubularins. The results reveal a number of potential signaling contexts for this family of enzymes, including an intriguing, novel role for myotubularin-related protein 3 and myotubularin-related protein 4 in the regulation of abscission, the final step of mitosis in which the membrane bridge remaining between two daughter cells is cleaved. Both depletion and overexpression of either myotubularin-related protein 3 or myotubularin-related protein 4 result in abnormal midbody morphology and cytokinesis failure. Interestingly, myotubularin-related protein 3 and myotubularin-related protein 4 do not exert their effects through lipid regulation at the midbody, but regulate abscission during early mitosis, by interacting with the mitotic kinase polo-like kinase 1, and with centrosomal protein of 55 kDa (CEP55), an important regulator of abscission. Structure-function analysis reveals that, consistent with known intramyotubularin interactions, myotubularin-related protein 3 and myotubularin-related protein 4 interact through their respective coiled coil domains. The interaction between myotubularin-related protein 3 and polo-like kinase 1 relies on the divergent, nonlipid binding Fab1, YOTB, Vac1, and EEA1 domain of myotubularin-related protein 3, and myotubularin-related protein 4 interacts with CEP55 through a short GPPXXXY motif, analogous to endosomal sorting complex required for transport-I components. Disruption of any of these interactions results in abscission failure, by disrupting the proper recruitment of CEP55, and subsequently, of endosomal sorting complex required for transport-I, to the midbody. Our data suggest that myotubularin-related protein 3 and myotubularin-related protein 4 may act as a bridge between CEP55 and polo-like kinase 1, ensuring proper CEP55 phosphorylation and regulating CEP55 recruitment to the midbody. This work provides a novel role for myotubularin-related protein 3/4 heterodimers, and highlights the temporal and spatial complexity of the regulation of cytokinesis.
Collapse
Affiliation(s)
- Nicole St-Denis
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Gagan D Gupta
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Zhen Yuan Lin
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Beatriz Gonzalez-Badillo
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Laurence Pelletier
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; §Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Anne-Claude Gingras
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; §Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada
| |
Collapse
|
135
|
Arquint C, Gabryjonczyk AM, Nigg EA. Centrosomes as signalling centres. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0464. [PMID: 25047618 DOI: 10.1098/rstb.2013.0464] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Centrosomes-as well as the related spindle pole bodies (SPBs) of yeast-have been extensively studied from the perspective of their microtubule-organizing roles. Moreover, the biogenesis and duplication of these organelles have been the subject of much attention, and the importance of centrosomes and the centriole-ciliary apparatus for human disease is well recognized. Much less developed is our understanding of another facet of centrosomes and SPBs, namely their possible role as signalling centres. Yet, many signalling components, including kinases and phosphatases, have been associated with centrosomes and spindle poles, giving rise to the hypothesis that these organelles might serve as hubs for the integration and coordination of signalling pathways. In this review, we discuss a number of selected studies that bear on this notion. We cover different processes (cell cycle control, development, DNA damage response) and organisms (yeast, invertebrates and vertebrates), but have made no attempt to be comprehensive. This field is still young and although the concept of centrosomes and SPBs as signalling centres is attractive, it remains primarily a concept-in need of further scrutiny. We hope that this review will stimulate thought and experimentation.
Collapse
Affiliation(s)
- Christian Arquint
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | | | - Erich A Nigg
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| |
Collapse
|
136
|
Tsai CY, Chen CH, Chang AYW, Chan JYH, Chan SHH. Upregulation of FLJ10540, a PI3K-association protein, in rostral ventrolateral medulla impairs brain stem cardiovascular regulation during mevinphos intoxication. Biochem Pharmacol 2015; 93:34-41. [PMID: 25449601 DOI: 10.1016/j.bcp.2014.10.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 12/15/2022]
Abstract
FLJ10540, originally identified as a microtubule-associated protein, induces cell proliferation and migration during tumorigenesis via the formation of FLJ10540-PI3K complex and enhancement of PI3K kinase activity. Interestingly, activation of PI3K/Akt cascade, leading to upregulation of nitric oxide synthase II (NOS II)/peroxynitrite signaling in the rostral ventrolateral medulla (RVLM), the brain stem site that maintains blood pressure and sympathetic vasomotor tone, mediates the impairment of brain stem cardiovascular regulation induced by the pesticide mevinphos. We evaluated the hypothesis that upregulation of FLJ10540 in the RVLM is upstream to this repertoire of signaling cascade that underpins mevinphos-induced circulatory depression. Microinjection bilaterally of mevinphos (10nmol) into the RVLM of anesthetized Sprague-Dawley rats induced a progressive hypotension that was accompanied by an increase (Phase I), followed by a decrease (Phase II) of an experimental index for baroreflex-mediated sympathetic vasomotor tone. There was augmentation in FLJ10540 mRNA in the RVLM or FLJ10540 protein in RVLM neurons, both of which were causally and temporally related to an augmentation of binding between the catalytic subunit (p110) and regulatory subunit (p85) of PI3K, phosphorylation of Akt at Thr308 site, and NOS II, superoxide or peroxynitrite level in the RVLM. Immunoneutralization of FJL10540 in the RVLM significantly antagonized those biochemical changes, and blunted the progressive hypotension and the reduced baroreflex-mediated sympathetic vasomotor tone during mevinphos intoxication. We conclude that upregulation of FLJ10540 in the RVLM elicits impairment of brain stem cardiovascular regulation that underpins circulatory depression during mevinphos intoxication via activation of PI3K/Akt/NOS II/peroxynitrite signaling cascade in the RVLM.
Collapse
Affiliation(s)
- Ching-Yi Tsai
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, Republic of China
| | - Chang-Han Chen
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, Republic of China
| | - Alice Y W Chang
- Institute of Physiology, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Julie Y H Chan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, Republic of China
| | - Samuel H H Chan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, Republic of China.
| |
Collapse
|
137
|
Zheng Y, Guo J, Li X, Xie Y, Hou M, Fu X, Dai S, Diao R, Miao Y, Ren J. An integrated overview of spatiotemporal organization and regulation in mitosis in terms of the proteins in the functional supercomplexes. Front Microbiol 2014; 5:573. [PMID: 25400627 PMCID: PMC4212687 DOI: 10.3389/fmicb.2014.00573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/11/2014] [Indexed: 12/22/2022] Open
Abstract
Eukaryotic cells may divide via the critical cellular process of cell division/mitosis, resulting in two daughter cells with the same genetic information. A large number of dedicated proteins are involved in this process and spatiotemporally assembled into three distinct super-complex structures/organelles, including the centrosome/spindle pole body, kinetochore/centromere and cleavage furrow/midbody/bud neck, so as to precisely modulate the cell division/mitosis events of chromosome alignment, chromosome segregation and cytokinesis in an orderly fashion. In recent years, many efforts have been made to identify the protein components and architecture of these subcellular organelles, aiming to uncover the organelle assembly pathways, determine the molecular mechanisms underlying the organelle functions, and thereby provide new therapeutic strategies for a variety of diseases. However, the organelles are highly dynamic structures, making it difficult to identify the entire components. Here, we review the current knowledge of the identified protein components governing the organization and functioning of organelles, especially in human and yeast cells, and discuss the multi-localized protein components mediating the communication between organelles during cell division.
Collapse
Affiliation(s)
- Yueyuan Zheng
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Junjie Guo
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Xu Li
- Orthopaedic Department of Anhui Medical University Affiliated Provincial Hospital Hefei, China
| | - Yubin Xie
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Mingming Hou
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Xuyang Fu
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Shengkun Dai
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Rucheng Diao
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Yanyan Miao
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Jian Ren
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| |
Collapse
|
138
|
Chen YJ, Lai KC, Kuo HH, Chow LP, Yih LH, Lee TC. HSP70 colocalizes with PLK1 at the centrosome and disturbs spindle dynamics in cells arrested in mitosis by arsenic trioxide. Arch Toxicol 2014; 88:1711-23. [PMID: 24623308 DOI: 10.1007/s00204-014-1222-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 03/03/2014] [Indexed: 11/26/2022]
Abstract
Heat shock protein 70 (HSP70) has been shown to be a substrate of Polo-like kinase 1 (PLK1), and it prevents cells arrested in mitosis by arsenic trioxide (ATO) from dying. Here, we report that HSP70 participates in ATO-induced spindle elongation, which interferes with mitosis progression. Our results demonstrate that HSP70 and PLK1 colocalize at the centrosome in ATO-arrested mitotic cells. HSP70 located at the centrosome was found to be phosphorylated by PLK1 at Ser⁶³¹ and Ser⁶³³. Moreover, unlike wild-type HSP70 (HSP70(wt)) and its phosphomimetic mutant (HSP70(SS631,633DD)), a phosphorylation-resistant mutant of HSP70 (HSP70(SS631,633AA)) failed to localize at the centrosome. ATO-induced spindle elongation was abolished in cells overexpressing HSP70(SS631,633AA). Conversely, mitotic spindles in cells ectopically expressing HSP70(SS631,633DD) were more resistant to nocodazole-induced depolymerization than in those expressing HSP70(wt) or HSP70(SS631,633AA). In addition, inhibition of PLK1 significantly reduced HSP70 phosphorylation and induced early onset of apoptosis in ATO-arrested mitotic cells. Taken together, our results indicate that PLK1-mediated phosphorylation and centrosomal localization of HSP70 may interfere with spindle dynamics and prevent apoptosis of ATO-arrested mitotic cells.
Collapse
Affiliation(s)
- Yu-Ju Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | | | | | | | | | | |
Collapse
|
139
|
Cashikar AG, Shim S, Roth R, Maldazys MR, Heuser JE, Hanson PI. Structure of cellular ESCRT-III spirals and their relationship to HIV budding. eLife 2014; 3. [PMID: 24878737 PMCID: PMC4073282 DOI: 10.7554/elife.02184] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/27/2014] [Indexed: 12/23/2022] Open
Abstract
The ESCRT machinery along with the AAA+ ATPase Vps4 drive membrane scission for trafficking into multivesicular bodies in the endocytic pathway and for the topologically related processes of viral budding and cytokinesis, but how they accomplish this remains unclear. Using deep-etch electron microscopy, we find that endogenous ESCRT-III filaments stabilized by depleting cells of Vps4 create uniform membrane-deforming conical spirals which are assemblies of specific ESCRT-III heteropolymers. To explore functional roles for ESCRT-III filaments, we examine HIV-1 Gag-mediated budding of virus-like particles and find that depleting Vps4 traps ESCRT-III filaments around nascent Gag assemblies. Interpolating between the observed structures suggests a new role for Vps4 in separating ESCRT-III from Gag or other cargo to allow centripetal growth of a neck constricting ESCRT-III spiral. DOI:http://dx.doi.org/10.7554/eLife.02184.001 Cells contain compartments called organelles that are enclosed within membranes similar to the plasma membrane that surrounds the cell itself. Cells police the proteins on their membranes and move old or damaged proteins into a type of organelle called an endosome. This involves the membrane folding in on itself to form a multivesicular body. The multivesicular bodies deliver their contents to organelles called lysosomes where the old proteins are destroyed. Although it is known that over 30 proteins are involved in the formation of multivesicular bodies, many aspects of how they operate are not well understood. Moreover, disruptions to this process contribute to a large number of diseases including forms of cancer and neurodegeneration. Importantly, the same proteins are hijacked by viruses such as HIV to help them escape from the cells they have infected. Most of the proteins involved in forming multivesicular bodies are part of the ESCRT (Endosomal Sorting Complex Required for Transport) system of proteins. A special set of these proteins—ESCRT-III—is thought to cut the membrane to release vesicles and viruses, as well as helping the membrane to deform. Previously, researchers have been unsure how the ESCRT-III complex works because it has a short lifespan and is too small to see on cellular membranes using standard techniques. Now Cashikar, Shim et al. have used a technique called deep-etch electron microscopy in combination with gene knockdown strategies to reveal the structure of the ESCRT-III complex inside cells. A protein called Vps4 is known to recycle ESCRT-III complexes, so Cashikar, Shim et al. studied cells in which the levels of Vps4 had been depleted in order to increase the lifespan of ESCRT-III complexes. In these cells filaments made of ESCRT-III complexes tended to form conical spirals that matched the size and shape of the vesicles and viruses ESCRT-III is thought to produce. ESCRT-III filaments also accumulated as rings around the molecules destined for incorporation into a vesicle or virus. This indicated a new role for Vps4: it separates ESCRT-III from the contents of the vesicle, leaving it free to form a spiral that drives release of the vesicle or virus from the cell. The next challenge will be to test the predictions of this model using techniques that can capture individual vesicle formation events in real time. Understanding the function of ESCRT-III in greater detail may suggest ways to manipulate this pathway to limit the replication of viruses or the degradation of membrane proteins. DOI:http://dx.doi.org/10.7554/eLife.02184.002
Collapse
Affiliation(s)
- Anil G Cashikar
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - Soomin Shim
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - Robyn Roth
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - Michael R Maldazys
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - John E Heuser
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - Phyllis I Hanson
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
140
|
Wu D, Asiedu M, Matsumura F, Wei Q. Phosphorylation of myosin II-interacting guanine nucleotide exchange factor (MyoGEF) at threonine 544 by aurora B kinase promotes the binding of polo-like kinase 1 to MyoGEF. J Biol Chem 2014; 289:7142-7150. [PMID: 24482237 PMCID: PMC3945374 DOI: 10.1074/jbc.m113.510388] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 01/24/2014] [Indexed: 01/09/2023] Open
Abstract
We previously reported that phosphorylation of myosin II-interacting guanine nucleotide exchange factor (MyoGEF) by polo-like kinase 1 (Plk1) promotes the localization of MyoGEF to the central spindle and increases MyoGEF activity toward RhoA during mitosis. In this study we report that aurora B-mediated phosphorylation of MyoGEF at Thr-544 creates a docking site for Plk1, leading to the localization and activation of MyoGEF at the central spindle. In vitro kinase assays show that aurora B can phosphorylate MyoGEF. T544A mutation drastically decreases aurora B-mediated phosphorylation of MyoGEF in vitro and in transfected HeLa cells. Coimmunoprecipitation and in vitro pulldown assays reveal that phosphorylation of MyoGEF at Thr-544 enhances the binding of Plk1 to MyoGEF. Immunofluorescence analysis shows that aurora B colocalizes with MyoGEF at the central spindle and midbody during cytokinesis. Suppression of aurora B activity by an aurora B inhibitor disrupts the localization of MyoGEF to the central spindle. In addition, T544A mutation interferes with the localization of MyoGEF to the cleavage furrow and decreases MyoGEF activity toward RhoA during mitosis. Taken together, our results suggest that aurora B coordinates with Plk1 to regulate MyoGEF activation and localization, thus contributing to the regulation of cytokinesis.
Collapse
Affiliation(s)
- Di Wu
- Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | - Michael Asiedu
- Department of Surgery, College of Medicine, Mayo Clinic Rochester, Minnesota 55902, and
| | - Fumio Matsumura
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08855
| | - Qize Wei
- Department of Biological Sciences, Fordham University, Bronx, New York 10458.
| |
Collapse
|
141
|
Green RA, Mayers JR, Wang S, Lewellyn L, Desai A, Audhya A, Oegema K. The midbody ring scaffolds the abscission machinery in the absence of midbody microtubules. ACTA ACUST UNITED AC 2014; 203:505-20. [PMID: 24217623 PMCID: PMC3824018 DOI: 10.1083/jcb.201306036] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The septins, but not midbody microtubules, are important for daughter cell cytoplasmic isolation and ESCRT-dependent midbody ring release during abscission. Abscission completes cytokinesis to form the two daughter cells. Although abscission could be organized from the inside out by the microtubule-based midbody or from the outside in by the contractile ring–derived midbody ring, it is assumed that midbody microtubules scaffold the abscission machinery. In this paper, we assess the contribution of midbody microtubules versus the midbody ring in the Caenorhabditis elegans embryo. We show that abscission occurs in two stages. First, the cytoplasm in the daughter cells becomes isolated, coincident with formation of the intercellular bridge; proper progression through this stage required the septins (a midbody ring component) but not the membrane-remodeling endosomal sorting complex required for transport (ESCRT) machinery. Second, the midbody and midbody ring are released into a specific daughter cell during the subsequent cell division; this stage required the septins and the ESCRT machinery. Surprisingly, midbody microtubules were dispensable for both stages. These results delineate distinct steps during abscission and highlight the central role of the midbody ring, rather than midbody microtubules, in their execution.
Collapse
Affiliation(s)
- Rebecca A Green
- Department of Cellular and Molecular Medicine, Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093
| | | | | | | | | | | | | |
Collapse
|
142
|
Tao J, Zhi X, Tian Y, Li Z, Zhu Y, Wang W, Xie K, Tang J, Zhang X, Wang L, Xu Z. CEP55 contributes to human gastric carcinoma by regulating cell proliferation. Tumour Biol 2014; 35:4389-99. [PMID: 24390615 DOI: 10.1007/s13277-013-1578-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 12/17/2013] [Indexed: 12/13/2022] Open
Abstract
Centrosomal protein 55 (CEP55) is the latest found member in the centrosomal relative protein family, which participates in cell-cycle regulation. CEP55 exists in many kinds of normal tissues and tumour cells such as hepatocellular carcinoma, and is important in carcinogenesis. However, the role of CEP55 in the pathogenesis of gastric cancer (GC) remains unclear. The mRNA levels of CEP55 in GC tissues and GC cell lines were examined by quantitative real-time PCR, and the protein expression of CEP55 in GC tissues was detected by Western blot and immunohistochemistry. The role of CEP55 in regulating the proliferation of GC cell lines was investigated both in vitro and in vivo. CEP55 was strongly upregulated in human GC, indicating that CEP55 contributed to carcinogenesis and progression of GC. Ectopic overexpression of CEP55 enhanced the cell proliferation, colony formation, and tumourigenicity of GC cells, whereas CEP55 knockdown inhibited these effects. We discovered that cell transformation induced by CEP55 was mediated by the AKT signalling pathway. Overexpression of CEP55 enhanced the phosphorylation of AKT and inhibited the activity of p21 WAF1/Cip1. In addition, cellular proliferation was suppressed as a result of cell cycle arrest at the G2/M phase in CEP55-knockdown cells. CEP55 expression was elevated in GC compared with normal control tissues. Credible evidence showed that CEP55 can be a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Jinqiu Tao
- Division of Gastric Surgery, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Le Bras S, Le Borgne R. Epithelial cell division – multiplying without losing touch. J Cell Sci 2014; 127:5127-37. [DOI: 10.1242/jcs.151472] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Epithelia are compact tissues comprising juxtaposed cells that function as mechanical and chemical barriers between the body and the environment. This barrier relies, in part, on adhesive contacts within adherens junctions, which are formed and stabilized by E-cadherin and catenin proteins linked to the actomyosin cytoskeleton. During development and throughout adult life, epithelia are continuously growing or regenerating, largely as a result of cell division. Although persistence of adherens junctions is needed for epithelial integrity, these junctions are continually remodelled during cell division. In this Commentary, we will focus on cytokinesis, the final step of mitosis, a multiparty phenomenon in which the adherens junction belt plays an essential role and during which a new cell–cell interface is generated between daughter cells. This new interface is the site of intense remodelling, where new adhesive contacts are assembled and cell polarity is transmitted from mother to daughter cells, ultimately becoming the site of cell signalling.
Collapse
|
144
|
Oncogenic fibulin-5 promotes nasopharyngeal carcinoma cell metastasis through the FLJ10540/AKT pathway and correlates with poor prognosis. PLoS One 2013; 8:e84218. [PMID: 24386352 PMCID: PMC3874010 DOI: 10.1371/journal.pone.0084218] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 11/13/2013] [Indexed: 12/21/2022] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is known for its high metastatic potential and locoregional recurrence, although the molecular alterations that are driving NPC metastasis remain unclear at this time. This study aimed to examine the expression of fibulin-5 in NPC, correlate the results with clinicopathological variables and survival, and to investigate the role of fibulin-5 in human NPC cell lines. Material and Methods Standard semi-quantitative-RT-PCR, quantitative-RT-PCR, immunoblotting, and immunohistochemistry were used to investigate the mRNA and protein expression profiles of fibulin-5 in normal and NPC tissues. Immunohistochemistry of fibulin-5 was correlated with clinicopathological characteristics by univariate analyses. NPC cells overexpressing fibulin-5 or fibulin-5-siRNA cells were generated by stable transfection to characterize the molecular mechanisms of fibulin-5-elicited cell growth and metastasis. Results Our results demonstrated that fibulin-5 overexpression in NPC specimens and significantly correlated with advanced tumor metastasis indicating a poor 5-year overall survival. Fibulin-5 was mainly expressed in the nucleus in human NPC specimens and cell lines. Functionally, fibulin-5 overexpression yielded fast growth in NPC cells. In addition, fibulin-5 promotes cell metastasis in NPC cells through increased FLJ10540 and phosphor-AKT activity. In contrast, siRNA depletion of fibulin-5 suppressed FLJ10540 expression and phosphor-AKT activity. Suppression of either fibulin-5 or FLJ10540 can cause significant inhibition with regards to cell motility in NPC cells. Finally, immunohistochemical analysis of human aggressive NPC specimens showed a significant and positive correlation between fibulin-5 and FLJ10540 expression. Conclusion Higher fibulin-5 expression is not only an important indicator of poor survival, but also contributes to the development of new therapeutic strategies in the FLJ10540/AKT pathway for NPC treatment.
Collapse
|
145
|
Harder LM, Bunkenborg J, Andersen JS. Inducing autophagy: a comparative phosphoproteomic study of the cellular response to ammonia and rapamycin. Autophagy 2013; 10:339-55. [PMID: 24300666 PMCID: PMC5396081 DOI: 10.4161/auto.26863] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Autophagy is a lysosomal-mediated catabolic process, which through degradation of different cytoplasmic components aids in maintaining cellular homeostasis and survival during exposure to extra- or intracellular stresses. Ammonia is a potential toxic and stress-inducing byproduct of glutamine catabolism, which has recently been found to induce autophagy in an MTOR independent way and support cancer cell survival. In this study, quantitative phosphoproteomics was applied to investigate the initial signaling events linking ammonia to the induction of autophagy. The MTOR inhibitor rapamycin was used as a reference treatment to emphasize the differences between an MTOR-dependent and -independent autophagy-induction. By this means 5901 phosphosites were identified of which 626 were treatment-specific regulated and 175 were coregulated. Investigation of the ammonia-specific regulated sites supported that MTOR activity was not affected, but indicated increased MAPK3 activity, regulation of proteins involved in Rho signal transduction, and a novel phosphorylation motif, serine-proline-threonine (SPT), which could be linked to cytoskeleton-associated proteins. MAPK3 could not be identified as the primary driver of ammonia-induced autophagy but instead the data suggested an upregulation of AMPK and the unfolded protein response (UPR), which might link ammonia to autophagy induction. Support of UPR induction was further obtained from the finding of increased protein levels of the ER stress markers DDIT3/CHOP and HSPA5 during ammonia treatment. The large-scale data set presented here comprises extensive high-quality quantitative information on phosphoprotein regulation in response to 2 very different autophagy inducers and should therefore be considered a general resource for the community.
Collapse
Affiliation(s)
- Lea M Harder
- Department of Biochemistry and Molecular Biology; University of Southern Denmark; Odense, Denmark
| | - Jakob Bunkenborg
- Department of Biochemistry and Molecular Biology; University of Southern Denmark; Odense, Denmark
| | - Jens S Andersen
- Department of Biochemistry and Molecular Biology; University of Southern Denmark; Odense, Denmark
| |
Collapse
|
146
|
Kumar A, Rajendran V, Sethumadhavan R, Purohit R. CEP proteins: the knights of centrosome dynasty. PROTOPLASMA 2013; 250:965-983. [PMID: 23456457 DOI: 10.1007/s00709-013-0488-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 02/12/2013] [Indexed: 06/01/2023]
Abstract
Centrosome forms the backbone of cell cycle progression mechanism. Recent debates have occurred regarding the essentiality of centrosome in cell cycle regulation. CEP family protein is the active component of centrosome and plays a vital role in centriole biogenesis and cell cycle progression control. A total of 31 proteins have been categorized into CEP family protein category and many more are under candidate evaluation. Furthermore, by the recent advancements in genomics and proteomics researches, several new CEP proteins have also been characterized. Here we have summarized the importance of CEP family proteins and their regulation mechanism involved in proper cell cycle progression. Further, we have reviewed the detailed molecular mechanism behind the associated pathological phenotypes and the possible therapeutic approaches. Proteins such as CEP57, CEP63, CEP152, CEP164, and CEP215 have been extensively studied with a detailed description of their molecular mechanisms, which are among the primary targets for drug discovery. Moreover, CEP27, CEP55, CEP70, CEP110, CEP120, CEP135, CEP192, CEP250, CEP290, and CEP350 also seem promising for future drug discovery approaches. Since the overview implicates that the overall researches on CEP proteins are not yet able to present significant details required for effective therapeutics development, thus, it is timely to discuss the importance of future investigations in this field.
Collapse
Affiliation(s)
- Ambuj Kumar
- Bioinformatics Division, School of Bio Sciences and Technology, Vellore Institute of Technology University, Vellore, 632014, Tamil Nadu, India
| | | | | | | |
Collapse
|
147
|
Nunes Bastos R, Gandhi SR, Baron RD, Gruneberg U, Nigg EA, Barr FA. Aurora B suppresses microtubule dynamics and limits central spindle size by locally activating KIF4A. J Cell Biol 2013; 202:605-21. [PMID: 23940115 PMCID: PMC3747307 DOI: 10.1083/jcb.201301094] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 07/12/2013] [Indexed: 02/07/2023] Open
Abstract
Anaphase central spindle formation is controlled by the microtubule-stabilizing factor PRC1 and the kinesin KIF4A. We show that an MKlp2-dependent pool of Aurora B at the central spindle, rather than global Aurora B activity, regulates KIF4A accumulation at the central spindle. KIF4A phosphorylation by Aurora B stimulates the maximal microtubule-dependent ATPase activity of KIF4A and promotes its interaction with PRC1. In the presence of phosphorylated KIF4A, microtubules grew more slowly and showed long pauses in growth, resulting in the generation of shorter PRC1-stabilized microtubule overlaps in vitro. Cells expressing only mutant forms of KIF4A lacking the Aurora B phosphorylation site overextended the anaphase central spindle, demonstrating that this regulation is crucial for microtubule length control in vivo. Aurora B therefore ensures that suppression of microtubule dynamic instability by KIF4A is restricted to a specific subset of microtubules and thereby contributes to central spindle size control in anaphase.
Collapse
Affiliation(s)
| | - Sapan R. Gandhi
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK
| | - Ryan D. Baron
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK
- Cancer Research Centre, University of Liverpool, Liverpool L3 9TA, England, UK
| | - Ulrike Gruneberg
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK
| | - Erich A. Nigg
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Francis A. Barr
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK
| |
Collapse
|
148
|
Telkoparan P, Erkek S, Yaman E, Alotaibi H, Bayık D, Tazebay UH. Coiled-coil domain containing protein 124 is a novel centrosome and midbody protein that interacts with the Ras-guanine nucleotide exchange factor 1B and is involved in cytokinesis. PLoS One 2013; 8:e69289. [PMID: 23894443 PMCID: PMC3716640 DOI: 10.1371/journal.pone.0069289] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/06/2013] [Indexed: 01/11/2023] Open
Abstract
Cytokinetic abscission is the cellular process leading to physical separation of two postmitotic sister cells by severing the intercellular bridge. The most noticeable structural component of the intercellular bridge is a transient organelle termed as midbody, localized at a central region marking the site of abscission. Despite its major role in completion of cytokinesis, our understanding of spatiotemporal regulation of midbody assembly is limited. Here, we report the first characterization of coiled-coil domain-containing protein-124 (Ccdc124), a eukaryotic protein conserved from fungi-to-man, which we identified as a novel centrosomal and midbody protein. Knockdown of Ccdc124 in human HeLa cells leads to accumulation of enlarged and multinucleated cells; however, centrosome maturation was not affected. We found that Ccdc124 interacts with the Ras-guanine nucleotide exchange factor 1B (RasGEF1B), establishing a functional link between cytokinesis and activation of localized Rap2 signaling at the midbody. Our data indicate that Ccdc124 is a novel factor operating both for proper progression of late cytokinetic stages in eukaryotes, and for establishment of Rap2 signaling dependent cellular functions proximal to the abscission site.
Collapse
Affiliation(s)
- Pelin Telkoparan
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Serap Erkek
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Elif Yaman
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Hani Alotaibi
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Defne Bayık
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Uygar H. Tazebay
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Gebze Institute of Technology, Kocaeli, Turkey
- * E-mail:
| |
Collapse
|
149
|
Sorting nexin 9 recruits clathrin heavy chain to the mitotic spindle for chromosome alignment and segregation. PLoS One 2013; 8:e68387. [PMID: 23861900 PMCID: PMC3702553 DOI: 10.1371/journal.pone.0068387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 06/02/2013] [Indexed: 12/22/2022] Open
Abstract
Sorting nexin 9 (SNX9) and clathrin heavy chain (CHC) each have roles in mitosis during metaphase. Since the two proteins directly interact for their other cellular function in endocytosis we investigated whether they also interact for metaphase and operate on the same pathway. We report that SNX9 and CHC functionally interact during metaphase in a specific molecular pathway that contributes to stabilization of mitotic spindle kinetochore (K)-fibres for chromosome alignment and segregation. This function is independent of their endocytic role. SNX9 residues in the clathrin-binding low complexity domain are required for CHC association and for targeting both CHC and transforming acidic coiled-coil protein 3 (TACC3) to the mitotic spindle. Mutation of these sites to serine increases the metaphase plate width, indicating inefficient chromosome congression. Therefore SNX9 and CHC function in the same molecular pathway for chromosome alignment and segregation, which is dependent on their direct association.
Collapse
|
150
|
LGALS3BP regulates centriole biogenesis and centrosome hypertrophy in cancer cells. Nat Commun 2013; 4:1531. [PMID: 23443559 DOI: 10.1038/ncomms2517] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 01/17/2013] [Indexed: 12/18/2022] Open
Abstract
Centrosome morphology and number are frequently deregulated in cancer cells. Here, to identify factors that are functionally relevant for centrosome abnormalities in cancer cells, we established a protein-interaction network around 23 centrosomal and cell-cycle regulatory proteins, selecting the interacting proteins that are deregulated in cancer for further studies. One of these components, LGALS3BP, is a centriole- and basal body-associated protein with a dual role, triggering centrosome hypertrophy when overexpressed and causing accumulation of centriolar substructures when downregulated. The cancer cell line SK-BR-3 that overexpresses LGALS3BP exhibits hypertrophic centrosomes, whereas in seminoma tissues with low expression of LGALS3BP, supernumerary centriole-like structures are present. Centrosome hypertrophy is reversed by depleting LGALS3BP in cells endogenously overexpressing this protein, supporting a direct role in centrosome aberration. We propose that LGALS3BP suppresses assembly of centriolar substructures, and when depleted, causes accumulation of centriolar complexes comprising CPAP, acetylated tubulin and centrin.
Collapse
|