1
|
Wang S, Lei Z, Liu W, Xiong J, Shi Y, Yang L, Gao Q, Le K, Zhang B. RCC2 promotes prostate cancer cell proliferation and migration through Hh/GLI1 signaling pathway and cancer stem-like cells. Biol Direct 2023; 18:80. [PMID: 38008751 PMCID: PMC10680210 DOI: 10.1186/s13062-023-00439-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Regulator of chromosome condensation 2 (RCC2) was a telophase disk-binding protein on mitosis, and functions as an oncogene in many human cancers. However, its role on prostate cancer (PCa) was unknown. The goal of this study is to explore the function of RCC 2 on PCa development. METHODS The expression of RCC2 and its methylation level, its correlation with lymph node metastasis or disease-free survival (DFS) was analyzed using TCGA database. The effect of RCC2 on PCa cell proliferation, migration and invasion were detected using CCK-8, cell colony formation, Transwell and wood healing assays. RNA-seq and GSEA analysis were used to search the downstream genes and pathways of RCC2 in mediated PCa progression. Western blot was used to detect the proteins in PCa cells transfected with indicated siRNAs or plasmids. RESULTS RCC2 had high expression and low promoter methylation level in PCa, and its expression was correlated with regional node metastasis and disease-free survival. Cell proliferation, migration, invasion and EMT of PCa cells in vitro were greatly enhanced after RCC2 overexpression, while the RCC2 knockdown suppressed these processes. RNA-seq and GSEA results showed the Hedgehog signaling regulator Gli1 and Gli3 were involved in RCC2 knockdown DU145 cells. Gli1 was also a marker of cancer stem-like cells (CSCs). Mechanistically, RCC2 induced cell growth, EMT, CSCs markers through Gli1; inhibiting Gli1 expression using siGli1 or GLI inhibitor suppressed cell progression in vitro and tumor growth in vivo. CONCLUSION In summary, RCC2 promoted PCa development through Hh/Gli1 signaling pathway via regulating EMT and CSCs.
Collapse
Affiliation(s)
- Shenghan Wang
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Zhentao Lei
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Wei Liu
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jie Xiong
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Yuqiang Shi
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Lin Yang
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Qiang Gao
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Kai Le
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Bao Zhang
- Department of Urology, Aerospace Center Hospital, No.15, Yuquan Road, Haidian District, Beijing, 100049, China.
| |
Collapse
|
2
|
Raut MS, Bansal S, Desai RS, Raut BS. A comparative immunohistochemical analysis of cortactin in orthokeratinized odontogenic cyst (OOC), sporadic odontogenic keratocyst (OKC), and syndromic OKC. J Oral Biosci 2021; 63:444-449. [PMID: 34534694 DOI: 10.1016/j.job.2021.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVES To evaluate and compare the immunohistochemical expression of cortactin in the epithelial lining of orthokeratinized odontogenic cyst (OOC), sporadic odontogenic keratocyst (OKC), and syndromic OKC. METHODS Formalin-fixed paraffin-embedded tissue blocks of histopathologically diagnosed cases of OOC, OKC, syndromic OKC, normal buccal mucosa (NBM), and oral squamous cell carcinoma (OSCC) were examined for immunohistochemical expression of cortactin. Clear brown cytoplasmic and membranous staining was considered positive. RESULTS A statistically significant difference was observed between OOC and syndromic OKC (p<0.001), as well as between sporadic OKC and syndromic OKC (p<0.001). Although not statistically significant, the expression of cortactin was slightly higher in the basal layer of NBM (mean=0.47), OOC (mean=0.27), sporadic OKC (mean=0.47) syndromic OKC (mean=1.53), and OSCC (mean=0.67) than in the parabasal layers of NBM (mean=0.27), OOC (mean=0.20), sporadic OKC (mean=0.47), syndromic OKC (mean=1.27), and OSCC (mean=0.60). CONCLUSION The expression of cortactin in the basal layer may suggest the formation of invadopodia in the basal layer where the invasion mechanism occurs. This finding is further supported by the higher localization of cortactin in areas of epithelial budding and daughter cysts in syndromic OKC, thereby reaffirming its possible association with recurrence.
Collapse
Affiliation(s)
- Mugdha S Raut
- Department of Oral Pathology and Microbiology, Nair Hospital Dental College, Dr. A.L Nair Road, Mumbai Central, Mumbai-400008, India.
| | - Shivani Bansal
- Department of Oral Pathology and Microbiology, Nair Hospital Dental College, Dr. A.L Nair Road, Mumbai Central, Mumbai-400008, India.
| | - Rajiv S Desai
- Department of Oral Pathology and Microbiology, Nair Hospital Dental College, Dr. A.L Nair Road, Mumbai Central, Mumbai-400008, India.
| | - Bhargav S Raut
- Furness General Hospital-Morecambe Bay Hospital Trust, Dalton Lane, Barrow in Furness, Cumbria, United Kingdom.
| |
Collapse
|
3
|
Calderon-Aparicio A, Bode AM. Roles of regulator of chromosome condensation 2 in cancer: Beyond its regulatory function in cell cycle. Oncol Rev 2021; 15:525. [PMID: 33824700 PMCID: PMC8018209 DOI: 10.4081/oncol.2021.525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/02/2021] [Indexed: 11/22/2022] Open
Abstract
Regulator of chromosome condensation 2 (RCC2) is an essential protein in order for mitosis to proceed properly. It localizes in the centrosome of chromosomes where is involved in chromosome segregation and cytokinesis. Furthermore, RCC2 associates with integrin networks at the plasma membrane where participates in the control of cell movement. Because of its known role in cell cycle, RCC2 has been linked with cancer progression. Several reports show that RCC2 induces cancer hallmarks, but the mechanisms explaining how RCC2 exerts these roles are widely unknown. Here, we aim to summarize the main findings explaining the roles and mechanisms of RCC2 in cancer promotion. RCC2 is overexpressed in different cancers, including glioblastoma, lung, ovarian, and esophageal which is related to proliferation, migration, invasion promotion in vitro and tumor progression and metastasis in vivo. Besides, RCC2 overexpression induces epithelial-mesenchymal transition and causes poorer prognosis in cancer patients. RCC2 overexpression has also been linked with resistance development to chemotherapy and radiotherapy by inhibiting apoptosis and activating cancer-promoting transcription factors. Unfortunately, not RCC2 inhibitors are currently available for further pre-clinical and clinical assays. Therefore, these findings emphasize the potential use of RCC2 as a targetable biomarker in cancer and highlight the importance for designing RCC2 chemical inhibitors to evaluate its efficacy in animal studies and clinical trials.
Collapse
Affiliation(s)
- Ali Calderon-Aparicio
- The Hormel Institute, University of Minnesota, Austin, MN.,Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD, USA
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN
| |
Collapse
|
4
|
Guo K, Zhao C, Lang B, Wang H, Zheng H, Zhang F. Regulator of Chromosome Condensation 2 Modulates Cell Cycle Progression, Tumorigenesis, and Therapeutic Resistance. Front Mol Biosci 2021; 7:620973. [PMID: 33521058 PMCID: PMC7838589 DOI: 10.3389/fmolb.2020.620973] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/08/2020] [Indexed: 01/03/2023] Open
Abstract
Accurate regulation of cell cycle is important for normal tissue development and homeostasis. RCC2 (Regulator of Chromosome Condensation 2) play a role as chromosomal passenger complex (CPC) implicated in all cell cycle phases. RCC2 was initially identified as Ran guanine exchange factor (GEF) for small G proteins. Therefore, RCC2 plays a key role in oncogenesis of most cancers. RCC2 is implicated in Colorectal Cancer (CRC), Lung Adenocarcinoma (LUAD), breast cancer, and ovarian cancer. Expression level of RCC2 protein determines regulation of tumor cell proliferation, invasion, metastasis, and radio-chemotherapeutic resistance. In this review, we explored proteins that interact with RCC2 to modulate tumor development and cancer therapeutic resistance by regulation of cell cycle process through various signaling pathways.
Collapse
Affiliation(s)
- Kun Guo
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Cheng Zhao
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Bin Lang
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Huiqin Wang
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Hang Zheng
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Feng Zhang
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| |
Collapse
|
5
|
RCC2 Expression Stimulates ER-Positive Breast Tumorigenesis. JOURNAL OF ONCOLOGY 2020; 2020:5619462. [PMID: 32565805 PMCID: PMC7262660 DOI: 10.1155/2020/5619462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/19/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023]
Abstract
Objective Regulator of chromosome condensation 2 (RCC2) has been reported to be involved in the regulation of cell cleavage. This study investigated the effect of RCC2 expression on breast tumorigenesis. Methods MCF-7 cells originating from estrogen receptor-positive (ER+) breast cancer were transfected with anti-RCC2 siRNA or RCC2-expressing plasmids. Cell proliferation, apoptosis, migration, and cytokine production in the transfected cells were examined using the CCK-8 assay, wound healing assay, and flow cytometry, respectively. PCR array was used to investigate the tumorigenic pathway of RCC2 in MCF-7 cells transfected with the anti-RCC2 siRNA. MCF-7 cells were also transfected with lentivirus-containing anti-RCC2 short hairpin RNA and were injected into BALB/c nude mice to generate tumor-bearing mice. Tumor growth in the mouse model was examined using magnetic resonance imaging by diffusion-weighted imaging analysis. Results Western blotting and immunohistochemistry detected significantly increased expression of RCC2 in ER + breast tumor tissues compared with breast fibroadenoma samples. Inhibiting RCC2 expression decreased cell migration and stimulated apoptosis in MCF-7 cells, while overexpressing RCC2 stimulated cell migration and inhibited apoptosis. The inhibition of RCC2 expression significantly decreased breast tumor growth and IL-6 levels in the tumor-bearing mice. PCR array demonstrated that inhibiting RCC2 expression significantly decreased the expression of IGF1 and TWIST1, two well-known tumor-enhancing genes, in MCF-7 cells; conversely, overexpressing RCC2 increased the expression levels of these two genes in the transfected cells. This result was verified in the mouse model following inhibition of RCC2 expression in MCF-7 cells. Additionally, estradiol-17β suppressed MCF-7 cell apoptosis, stimulated cell proliferation and cell migration, and increased RCC2, IGF1, and TWIST1 expression. The siRNA-mediated inhibition of RCC2 expression alleviated the inhibitory effects of estrogen on apoptosis in MCF-7 cells, while overexpressing RCC2 enhanced the estrogen-driven inhibition of apoptosis. Modifying RCC2 expression had no impact on MCF-7 cell proliferation in the presence or absence of estradiol-17β. Conclusions Our results suggest that estrogen-induced RCC2 expression prompts IGF1, TWIST1, and IL-6 expression, stimulates cell migration, and inhibits apoptosis to contribute to ER + breast tumorigenesis.
Collapse
|
6
|
Pang B, Wu N, Guan R, Pang L, Li X, Li S, Tang L, Guo Y, Chen J, Sun D, Sun H, Dai J, Bai J, Ji G, Liu P, Liu A, Wang Q, Xiao S, Fu S, Jin Y. Overexpression of RCC2 Enhances Cell Motility and Promotes Tumor Metastasis in Lung Adenocarcinoma by Inducing Epithelial-Mesenchymal Transition. Clin Cancer Res 2017; 23:5598-5610. [PMID: 28606921 DOI: 10.1158/1078-0432.ccr-16-2909] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 04/25/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Investigate the role of regulator of chromosome condensation 2 (RCC2) on lung adenocarcinoma (LUAD) metastasis.Experimental Design: Clinical specimens were used to assess the impact of RCC2 on LUAD metastasis. Mouse models, cytobiology, and molecular biology assays were performed to elucidate the function and underlying mechanisms of RCC2 in LUAD.Results: RCC2 expression was frequently increased in LUADs (88/122, 72.13%). It was confirmed by analysis of a larger cohort of TCGA RNA-seq data containing 488 LUADs and 58 normal lung tissues (P < 0.001). Importantly, increased level of RCC2 was significantly associated with T status of tumor (P = 0.002), lymph node metastasis (P = 0.004), and advanced clinical stage (P = 0.001). Patients with LUAD with higher expression of RCC2 had shorter overall survival. Cox regression analysis demonstrated that RCC2 was an independent poorer prognostic factor for patients with LUAD. Moreover, forced expression of RCC2 promoted intrapulmonary metastasis in vivo and significantly enhanced LUAD cell migration, invasion, and proliferation in vitro Further study found that RCC2 induced epithelial-mesenchymal transition (EMT) and also stimulated the expression of MMP-2 and MMP-9. In addition, RCC2 was able to activate JNK, while inhibition of JNK suppressed the effect of RCC2 on LUAD cell migration, invasion, EMT, and the expression of MMP-2 and MMP-9.Conclusions: RCC2 plays a pivotal role in LUAD metastasis by inducing EMT via activation of MAPK-JNK signaling. Clin Cancer Res; 23(18); 5598-610. ©2017 AACR.
Collapse
Affiliation(s)
- Bo Pang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Nan Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Rongwei Guan
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Lin Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinlei Li
- Department of Human Anatomy, Harbin Medical University, Harbin, China
| | - Su Li
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Liudi Tang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Ying Guo
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jialei Chen
- Department of Thoracic Surgery, The Second Affiliated Clinical Hospital, Harbin Medical University, Harbin, China
| | - Donglin Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Haiming Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jialin Dai
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jing Bai
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Guohua Ji
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Peng Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - An Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Qiushi Wang
- Department of Thoracic Surgery, The Second Affiliated Clinical Hospital, Harbin Medical University, Harbin, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Songbin Fu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China. .,Key Laboratory of Medical Genetics (Harbin Medical University), Heilongjiang Higher Education Institutions, Harbin, China
| | - Yan Jin
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China. .,Key Laboratory of Medical Genetics (Harbin Medical University), Heilongjiang Higher Education Institutions, Harbin, China
| |
Collapse
|
7
|
Analysis of Microarray Data on Gene Expression and Methylation to Identify Long Non-coding RNAs in Non-small Cell Lung Cancer. Sci Rep 2016; 6:37233. [PMID: 27849024 PMCID: PMC5110979 DOI: 10.1038/srep37233] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/26/2016] [Indexed: 12/28/2022] Open
Abstract
To identify what long non-coding RNAs (lncRNAs) are involved in non-small cell lung cancer (NSCLC), we analyzed microarray data on gene expression and methylation. Gene expression chip and HumanMethylation450BeadChip were used to interrogate genome-wide expression and methylation in tumor samples. Differential expression and methylation were analyzed through comparing tumors with adjacent non-tumor tissues. LncRNAs expressed differentially and correlated with coding genes and DNA methylation were validated in additional tumor samples using RT-qPCR and pyrosequencing. In vitro experiments were performed to evaluate lncRNA’s effects on tumor cells. We identified 8,500 lncRNAs expressed differentially between tumor and non-tumor tissues, of which 1,504 were correlated with mRNA expression. Two of the lncRNAs, LOC146880 and ENST00000439577, were positively correlated with expression of two cancer-related genes, KPNA2 and RCC2, respectively. High expression of LOC146880 and ENST00000439577 were also associated with poor survival. Analysis of lncRNA expression in relation to DNA methylation showed that LOC146880 expression was down-regulated by DNA methylation in its promoter. Lowering the expression of LOC146880 or ENST00000439577 in tumor cells could inhibit cell proliferation, invasion and migration. Analysis of microarray data on gene expression and methylation allows us to identify two lncRNAs, LOC146880 and ENST00000439577, which may promote the progression of NSCLC.
Collapse
|
8
|
Alblazi KMO, Siar CH. Cellular protrusions--lamellipodia, filopodia, invadopodia and podosomes--and their roles in progression of orofacial tumours: current understanding. Asian Pac J Cancer Prev 2016; 16:2187-91. [PMID: 25824735 DOI: 10.7314/apjcp.2015.16.6.2187] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Protrusive structures formed by migrating and invading cells are termed lamellipodia, filopodia, invadopodia and podosomes. Lamellipodia and filopodia appear on the leading edges of migrating cells and function to command the direction of the migrating cells. Invadopodia and podosomes are special F-actin-rich matrix-degrading structures that arise on the ventral surface of the cell membrane. Invadopodia are found in a variety of carcinomatous cells including squamous cell carcinoma of head and neck region whereas podosomes are found in normal highly motile cells of mesenchymal and myelomonocytic lineage. Invadopodia-associated protein markers consisted of 129 proteins belonging to different functional classes including WASP, NWASP, cortactin, Src kinase, Arp 2/3 complex, MT1-MMP and F-actin. To date, our current understanding on the role(s) of these regulators of actin dynamics in tumors of the orofacial region indicates that upregulation of these proteins promotes invasion and metastasis in oral squamous cell carcinoma, is associated with poor/worst prognostic outcome in laryngeal cancers, contributes to the persistent growth and metastasis characteristics of salivary gland adenoid cystic carcinoma, is a significant predictor of increased cancer risk in oral mucosal premalignant lesions and enhances local invasiveness in jawbone ameloblastomas.
Collapse
Affiliation(s)
- Kamila Mohamed Om Alblazi
- Department of Oro-Maxillofacial Surgical and Medical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia E-mail :
| | | |
Collapse
|
9
|
Yu C, Guo J, Liu Y, Jia J, Jia R, Fan M. Oral squamous cancer cell exploits hnRNP A1 to regulate cell cycle and proliferation. J Cell Physiol 2015; 230:2252-61. [PMID: 25752295 DOI: 10.1002/jcp.24956] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 02/05/2015] [Indexed: 01/08/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is a common human malignant tumor with high mortality. So far, the molecular pathogenesis of OSCC remains largely unclear. Heterogeneous nuclear ribonucleoprotein (hnRNP) A1 is an important multi-function splicing factor and closely related to tumorigenesis. hnRNP A1 is overexpressed in various tumors, and promotes aerobic glycolysis and elongation of telomere, but the function of hnRNP A1 in cell cycle and proliferation remains unclear. We found that hnRNP A1 was overexpressed in OSCC tissues, and was required for the growth of OSCC cells. Moreover, hnRNP A1 was highly expressed in the G2/M cell cycle phase. Knockdown of hnRNP A1 induced G2/M arrest. DNA microarray assay result showed that hnRNP A1 regulated the expression of a number of target genes associated with G2/M phase. Moreover, hnRNP A1 controlled the alternative splicing of CDK2 exon 5. These findings suggested that hnRNP A1 plays key roles in the regulation of cell cycle progression and pathogenesis of OSCC.
Collapse
Affiliation(s)
- Cheng Yu
- Hubei-MOST KLOS & KLOBME, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Jihua Guo
- Hubei-MOST KLOS & KLOBME, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Yu Liu
- College of Life Sciences, Wuhan University, Wuhan, PR China
| | - Jun Jia
- Hubei-MOST KLOS & KLOBME, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Rong Jia
- Hubei-MOST KLOS & KLOBME, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Mingwen Fan
- Hubei-MOST KLOS & KLOBME, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| |
Collapse
|
10
|
Papini D, Langemeyer L, Abad MA, Kerr A, Samejima I, Eyers PA, Jeyaprakash AA, Higgins JMG, Barr FA, Earnshaw WC. TD-60 links RalA GTPase function to the CPC in mitosis. Nat Commun 2015; 6:7678. [PMID: 26158537 PMCID: PMC4510650 DOI: 10.1038/ncomms8678] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 05/29/2015] [Indexed: 12/26/2022] Open
Abstract
TD-60 (also known as RCC2) is a highly conserved protein that structurally resembles the Ran guanine exchange factor (GEF) RCC1, but has not previously been shown to have GEF activity. TD-60 has a typical chromosomal passenger complex (CPC) distribution in mitotic cells, but associates with integrin complexes and is involved in cell motility during interphase. Here we show that TD-60 exhibits GEF activity, in vitro and in cells, for the small GTPase RalA. TD-60 or RalA depletion causes spindle abnormalities in prometaphase associated with abnormal centromeric accumulation of CPC components. TD-60 and RalA apparently work together to contribute to the regulation of kinetochore-microtubule interactions in early mitosis. Importantly, several mitotic phenotypes caused by TD-60 depletion are reverted by the expression of a GTP-locked mutant, RalA (Q72L). The demonstration that a small GTPase participates in the regulation of the CPC reveals a level of mitotic regulation not suspected in previous studies.
Collapse
Affiliation(s)
- Diana Papini
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
- Institute for Cell and Molecular Biosciences (ICaMB), Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Lars Langemeyer
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Maria A. Abad
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Alastair Kerr
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Itaru Samejima
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Patrick A. Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK
| | - A. Arockia Jeyaprakash
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Jonathan M. G. Higgins
- Institute for Cell and Molecular Biosciences (ICaMB), Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Francis A. Barr
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - William C. Earnshaw
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| |
Collapse
|
11
|
Bruun J, Kolberg M, Ahlquist TC, Røyrvik EC, Nome T, Leithe E, Lind GE, Merok MA, Rognum TO, Bjørkøy G, Johansen T, Lindblom A, Sun XF, Svindland A, Liestøl K, Nesbakken A, Skotheim RI, Lothe RA. Regulator of Chromosome Condensation 2 Identifies High-Risk Patients within Both Major Phenotypes of Colorectal Cancer. Clin Cancer Res 2015; 21:3759-70. [PMID: 25910952 DOI: 10.1158/1078-0432.ccr-14-3294] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/29/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Colorectal cancer has high incidence and mortality worldwide. Patients with microsatellite instable (MSI) tumors have significantly better prognosis than patients with microsatellite stable (MSS) tumors. Considerable variation in disease outcome remains a challenge within each subgroup, and our purpose was to identify biomarkers that improve prediction of colorectal cancer prognosis. EXPERIMENTAL DESIGN Mutation analyses of 42 MSI target genes were performed in two independent MSI tumor series (n = 209). Markers that were significantly associated with prognosis in the test series were assessed in the validation series, followed by functional and genetic explorations. The clinical potential was further investigated by immunohistochemistry in a population-based colorectal cancer series (n = 903). RESULTS We identified the cell-cycle gene regulator of chromosome condensation 2 (RCC2) as a cancer biomarker. We found a mutation in the 5' UTR region of RCC2 that in univariate and multivariate analyses was significantly associated with improved outcome in the MSI group. This mutation caused reduction of protein expression in dual luciferase gene reporter assays. siRNA knockdown in MSI colon cancer cells (HCT15) caused reduced cell proliferation, cell-cycle arrest, and increased apoptosis. Massive parallel sequencing revealed few RCC2 mutations in MSS tumors. However, weak RCC2 protein expression was significantly associated with poor prognosis, independent of clinical high-risk parameters, and stratifies clinically important patient subgroups with MSS tumors, including elderly patients (>75 years), stage II patients, and those with rectal cancer. CONCLUSIONS Impaired RCC2 affects functional and clinical endpoints of colorectal cancer. High-risk patients with either MSI or MSS tumors can be identified with cost-effective routine RCC2 assays.
Collapse
Affiliation(s)
- Jarle Bruun
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Matthias Kolberg
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Terje C Ahlquist
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Ellen C Røyrvik
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. Department of Molecular Biosciences, University of Oslo, Oslo, Norway. Department of Oncology, University of Oxford, ORCRB, Headington, Oxford, United Kingdom
| | - Torfinn Nome
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Edward Leithe
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Guro E Lind
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marianne A Merok
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway. Department of Gastrointestinal Surgery, Aker Hospital-Oslo University Hospital, Oslo, Norway
| | - Torleiv O Rognum
- Faculty of Medicine, University of Oslo, Norway. Division of Forensic Medicine, Department of Forensic Pathology and Clinical Forensic Medicine, the Norwegian Institute of Public Health, Oslo, Norway
| | - Geir Bjørkøy
- University College of Sør-Trøndelag, Trondheim, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Xiao-Feng Sun
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, County Council of Östergötland, University of Linköping, Linköping, Sweden
| | - Aud Svindland
- Faculty of Medicine, University of Oslo, Norway. Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Knut Liestøl
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway. Department of Informatics, Faculty of Mathematics and Natural Sciences, Oslo, Norway
| | - Arild Nesbakken
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway. Department of Gastrointestinal Surgery, Aker Hospital-Oslo University Hospital, Oslo, Norway. Faculty of Medicine, University of Oslo, Norway
| | - Rolf I Skotheim
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway. Department of Informatics, Faculty of Mathematics and Natural Sciences, Oslo, Norway
| | - Ragnhild A Lothe
- Department for Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway. Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway. Department of Molecular Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
12
|
Havrylov S, Park M. MS/MS-based strategies for proteomic profiling of invasive cell structures. Proteomics 2014; 15:272-86. [PMID: 25303514 DOI: 10.1002/pmic.201400220] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/19/2014] [Accepted: 10/01/2014] [Indexed: 12/29/2022]
Abstract
Acquired capacity of cancer cells to penetrate through the extracellular matrix of surrounding tissues is a prerequisite for tumour metastatic spread - the main source of cancer-associated mortality. Through combined efforts of many research groups, we are beginning to understand that the ability of cells to invade through the extracellular matrix is a multi-faceted phenomenon supported by variety of specialised protrusive cellular structures, primarily pseudopodia, invadopodia and podosomes. Additionally, secreted extracellular vesicles are being increasingly recognised as important mediators of invasive cell phenotypes and therefore may be considered bona fide invasive cell structures. Dissection of the molecular makings underlying biogenesis and function of all of these structures is crucial to identify novel targets for specific anti-metastatic therapies. Rapid advances and growing accessibility of MS/MS-based protein identification made this family of techniques a suitable and appropriate choice for proteomic profiling of invasive cell structures. In this review, we provide a summary of current progress in the characterisation of protein composition and topology of protein interaction networks of pseudopodia, invadopodia, podosomes and extracellular vesicles, as well as outline challenges and perspectives of the field.
Collapse
Affiliation(s)
- Serhiy Havrylov
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada; Department of Medicine, McGill University, Montreal, QC, Canada
| | | |
Collapse
|
13
|
Aurora A kinase modulates actin cytoskeleton through phosphorylation of Cofilin: Implication in the mitotic process. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2719-29. [PMID: 25090971 DOI: 10.1016/j.bbamcr.2014.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 11/22/2022]
Abstract
Aurora A kinase regulates early mitotic events through phosphorylation and activation of a variety of proteins. Specifically, Aur-A is involved in centrosomal separation and formation of mitotic spindles in early prophase. The effect of Aur-A on mitotic spindles is mediated by the modulation of microtubule dynamics and association with microtubule binding proteins. In this study we show that Aur-A exerts its effects on spindle organization through the regulation of the actin cytoskeleton. Aurora A phosphorylates Cofilin at multiple sites including S(3) resulting in the inactivation of its actin depolymerizing function. Aur-A interacts with Cofilin in early mitotic phases and regulates its phosphorylation status. Cofilin phosphorylation follows a dynamic pattern during the progression of prophase to metaphase. Inhibition of Aur-A activity induced a delay in the progression of prophase to metaphase. Aur-A inhibitor also disturbed the pattern of Cofilin phosphorylation, which correlated with the mitotic delay. Our results establish a novel function of Aur-A in the regulation of actin cytoskeleton reorganization, through Cofilin phosphorylation during early mitotic stages.
Collapse
|
14
|
Gerrits H, Paradé MCBC, Koonen-Reemst AMCB, Bakker NEC, Timmer-Hellings L, Sollewijn Gelpke MD, Gossen JA. Reversible infertility in a liver receptor homologue-1 (LRH-1)-knockdown mouse model. Reprod Fertil Dev 2014; 26:293-306. [DOI: 10.1071/rd12131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 12/22/2012] [Indexed: 12/15/2022] Open
Abstract
Liver receptor homologue-1 (LRH-1) is an orphan nuclear receptor that has been implicated in steroid hormone biosynthesis and fertility. Herein we describe a transgenic inducible short hairpin (sh) RNA mouse model that was used to study the effect of transient LRH-1 knockdown in vivo. Induction of expression of the shRNA directed against LRH-1 for 2–6 weeks resulted in 80% knockdown of LRH-1 protein in the ovary and complete infertility. Gonadotropin hyperstimulation could not rescue the observed defects in ovulation and corpus luteum formation in LRH-1-knockdown mice. The infertility phenotype was fully reversible because LRH-1-knockdown females became pregnant and delivered normal size litters and healthy pups after cessation of LRH-1 shRNA expression. Timed ovarian microarray analysis showed that, in line with the observed decrease in plasma progesterone levels, key steroid biosynthesis genes, namely Star, Cyp11a1, Hsd3b and Scarb1, were downregulated in LRH-1-knockdown ovaries. In contrast with what has been described previously, no clear effect was observed on oestrogenic activity in LRH-1-knockdown mice. Only Sult1e1 and, surprisingly, Hsd17b7 expression was modulated with potentially opposite effects on oestradiol bioavailability. In conclusion, the fully reversible infertility phenotype of LRH-1-knockdown mice shows the feasibility of an LRH-1 antagonist as new contraceptive therapy with a mechanism of action that most prominently affects cholesterol availability and progesterone production.
Collapse
|
15
|
Yenjerla M, Panopoulos A, Reynaud C, Fotedar R, Margolis RL. TD-60 is required for interphase cell cycle progression. Cell Cycle 2013; 12:837-41. [PMID: 23388455 DOI: 10.4161/cc.23821] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We previously identified TD-60 (RCC2) as a mitotic centromere-associated protein that is necessary for proper completion of mitosis. We now report that TD-60 is an essential regulator of cell cycle progression during interphase. siRNA suppression blocks progression of mammalian G₁/S phase cells and progression of G₂ cells into mitosis. Prolonged arrest occurs both in non-transformed cells and in transformed cells lacking functional p53. TD-60 associates with Rac1 and Arf6 and has recently been demonstrated to be an element of α5β1 integrin and cortactin interactomes. These associations with known elements of cell cycle control, together with our data, suggest that TD-60 is an essential component of one or more signaling pathways that drive cell cycle progression. During mitosis, TD-60 is required for correct assembly of the mitotic spindle and activation of key mitotic proteins. In contrast, in interphase TD-60 promotes cell cycle progression through what must be distinct mechanisms. TD-60 thus appears to be one of the growing categories of proteins that "moonlight," or have more than one distinct cellular function.
Collapse
Affiliation(s)
- Mythili Yenjerla
- Tumor Development Program, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
16
|
Byron A, Humphries JD, Humphries MJ. Alternative cellular roles for proteins identified using proteomics. J Proteomics 2012; 75:4184-5. [PMID: 22579753 PMCID: PMC4234028 DOI: 10.1016/j.jprot.2012.04.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 04/30/2012] [Indexed: 10/28/2022]
Affiliation(s)
- Adam Byron
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Jonathan D. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Martin J. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
17
|
Matrix rigidity regulates cancer cell growth by modulating cellular metabolism and protein synthesis. PLoS One 2012; 7:e37231. [PMID: 22623999 PMCID: PMC3356407 DOI: 10.1371/journal.pone.0037231] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 04/16/2012] [Indexed: 11/19/2022] Open
Abstract
Background Tumor cells in vivo encounter diverse types of microenvironments both at the site of the primary tumor and at sites of distant metastases. Understanding how the various mechanical properties of these microenvironments affect the biology of tumor cells during disease progression is critical in identifying molecular targets for cancer therapy. Methodology/Principal Findings This study uses flexible polyacrylamide gels as substrates for cell growth in conjunction with a novel proteomic approach to identify the properties of rigidity-dependent cancer cell lines that contribute to their differential growth on soft and rigid substrates. Compared to cells growing on more rigid/stiff substrates (>10,000 Pa), cells on soft substrates (150–300 Pa) exhibited a longer cell cycle, due predominantly to an extension of the G1 phase of the cell cycle, and were metabolically less active, showing decreased levels of intracellular ATP and a marked reduction in protein synthesis. Using stable isotope labeling of amino acids in culture (SILAC) and mass spectrometry, we measured the rates of protein synthesis of over 1200 cellular proteins under growth conditions on soft and rigid/stiff substrates. We identified cellular proteins whose syntheses were either preferentially inhibited or preserved on soft matrices. The former category included proteins that regulate cytoskeletal structures (e.g., tubulins) and glycolysis (e.g., phosphofructokinase-1), whereas the latter category included proteins that regulate key metabolic pathways required for survival, e.g., nicotinamide phosphoribosyltransferase, a regulator of the NAD salvage pathway. Conclusions/Significance The cellular properties of rigidity-dependent cancer cells growing on soft matrices are reminiscent of the properties of dormant cancer cells, e.g., slow growth rate and reduced metabolism. We suggest that the use of relatively soft gels as cell culture substrates would allow molecular pathways to be studied under conditions that reflect the different mechanical environments encountered by cancer cells upon metastasis to distant sites.
Collapse
|