1
|
Dong W, Li X, Cheng L, Yang J, Zhao Z, Qiang X, Li P, Wu J, Guo L. RAE1 promotes gastric carcinogenesis and epithelial-mesenchymal transition. Arch Biochem Biophys 2024; 754:109896. [PMID: 38417691 DOI: 10.1016/j.abb.2024.109896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 03/01/2024]
Abstract
AIMS The purpose of this study was to explore the role of RAE1 in the invasion and metastasis of gastric cancer (GC) cells. MATERIALS AND METHODS RAE1 expression in GC cells was determined by reverse-transcription polymerase chain reaction (qRT-PCR) and Western blotting (WB). Cell models featuring RAE1 gene silencing and overexpression were constructed by lentiviral transfection; The proliferation, migration, and invasion ability of cells were detected by cell counting, colony formation assay, would healing assay, and transwell invasion and migration test. WB analysis of ERK/MAPK signaling pathway (ERK1/2, p-ERK1/2, c-Myc) and EMT-related molecules (ZEB1, E-cadherin, N-cadherin, and Vimentin). RESULTS The expression level of RAE1 in GC was notably higher than in adjacent tissues. Elevated RAE1 expression correlated with an unfavorable prognosis for GC patients. Knockdown of RAE1, as compared to the control group, resulted in a significant inhibition of proliferation, migration, and invasion abilities in GC cell lines. Furthermore, RAE1 knockdown led to a substantial decrease in the expression of N-cadherin, vimentin, ZEB1, p-ERK1/2, and c-Myc proteins, coupled with a marked increase in E-cadherin expression. The biological effects of RAE1 in GC cells were effectively reversed by the inhibition of the ERK/MAPK signaling pathway using SCH772984. Additionally, RAE1 knockdown demonstrated a suppressive effect on GC tumor size in vivo. Immunohistochemistry (IHC) results revealed significantly lower expression of Ki-67 in RAE1 knockout mice compared to the control group. CONCLUSIONS RAE1 promotes GC cell migration and invasion through the ERK/MAPK pathway and is a potential therapeutic target for GC therapy.
Collapse
Affiliation(s)
- Wenhui Dong
- Department of Gastroenterology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xiaofei Li
- Department of Gastroenterology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Lulu Cheng
- Department of Gastroenterology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jing Yang
- Department of Pathology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ziyan Zhao
- Department of Hematology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xihui Qiang
- Department of Gastroenterology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Pengmei Li
- Department of Gastroenterology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ju Wu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.
| | - Lianyi Guo
- Department of Gastroenterology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
2
|
Xu J, Wu F, Zhu Y, Wu T, Cao T, Gao W, Liu M, Qian W, Feng G, Xi X, Hou S. ANGPTL4 regulates ovarian cancer progression by activating the ERK1/2 pathway. Cancer Cell Int 2024; 24:54. [PMID: 38311733 PMCID: PMC10838463 DOI: 10.1186/s12935-024-03246-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 01/25/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) has the highest mortality rate among all gynecological malignancies. A hypoxic microenvironment is a common feature of solid tumors, including ovarian cancer, and an important driving factor of tumor cell survival and chemo- and radiotherapy resistance. Previous research identified the hypoxia-associated gene angiopoietin-like 4 (ANGPTL4) as both a pro-angiogenic and pro-metastatic factor in tumors. Hence, this work aimed to further elucidate the contribution of ANGPTL4 to OC progression. METHODS The expression of hypoxia-associated ANGPTL4 in human ovarian cancer was examined by bioinformatics analysis of TCGA and GEO datasets. The CIBERSORT tool was used to analyze the distribution of tumor-infiltrating immune cells in ovarian cancer cases in TCGA. The effect of ANGPTL4 silencing and overexpression on the proliferation and migration of OVCAR3 and A2780 OC cells was studied in vitro, using CCK-8, colony formation, and Transwell assays, and in vivo, through subcutaneous tumorigenesis assays in nude mice. GO enrichment analysis and WGCNA were performed to explore biological processes and genetic networks associated with ANGPTL4. The results obtained were corroborated in OC cells in vitro by western blotting. RESULTS Screening of hypoxia-associated genes in OC-related TCGA and GEO datasets revealed a significant negative association between ANGPTL4 expression and patient survival. Based on CIBERSORT analysis, differential representation of 14 distinct tumor-infiltrating immune cell types was detected between low- and high-risk patient groups. Silencing of ANGPTL4 inhibited OVCAR3 and A2780 cell proliferation and migration in vitro and reduced the growth rate of xenografted OVCAR3 cells in vivo. Based on results from WGCNA and previous studies, western blot assays in cultured OC cells demonstrated that ANGPTL4 activates the Extracellular signal-related kinases 1 and 2 (ERK1/2) pathway and this results in upregulation of c-Myc, Cyclin D1, and MMP2 expression. Suggesting that the above mechanism mediates the pro-oncogenic actions of ANGPTL4T in OC, the pro-survival effects of ANGPTL4 were largely abolished upon inhibition of ERK1/2 signaling with PD98059. CONCLUSIONS Our work suggests that the hypoxia-associated gene ANGPTL4 stimulates OC progression through activation of the ERK1/2 pathway. These findings may offer a new prospect for targeted therapies for the treatment of OC.
Collapse
Affiliation(s)
- Jiaqi Xu
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China
| | - Fei Wu
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China
| | - Yue Zhu
- Department of Breast and Thyroid Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China
| | - Tiantian Wu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Tianyue Cao
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China
| | - Wenxin Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Meng Liu
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China
| | - Weifeng Qian
- Department of Breast and Thyroid Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China
| | - Guannan Feng
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China
| | - Xiaoxue Xi
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China.
| | - Shunyu Hou
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, No.26, Daoqian Street, Suzhou, 215002, Jiangsu, China.
| |
Collapse
|
3
|
Ghozlan H, Showalter A, Lee E, Zhu X, Khaled AR. Chaperonin-Containing TCP1 Complex (CCT) Promotes Breast Cancer Growth Through Correlations With Key Cell Cycle Regulators. Front Oncol 2021; 11:663877. [PMID: 33996588 PMCID: PMC8121004 DOI: 10.3389/fonc.2021.663877] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Uncontrolled proliferation as a result of dysregulated cell cycling is one of the hallmarks of cancer. Therapeutically targeting pathways that control the cell cycle would improve patient outcomes. However, the development of drug resistance and a limited number of inhibitors that target multiple cell cycle modulators are challenges that impede stopping the deregulated growth that leads to malignancy. To advance the discovery of new druggable targets for cell cycle inhibition, we investigated the role of Chaperonin-Containing TCP1 (CCT or TRiC) in breast cancer cells. CCT, a type II chaperonin, is a multi-subunit protein-folding complex that interacts with many oncoproteins and mutant tumor suppressors. CCT subunits are highly expressed in a number of cancers, including breast cancer. We found that expression of one of the CCT subunits, CCT2, inversely correlates with breast cancer patient survival and is subject to copy number alterations through genomic amplification. To investigate a role for CCT2 in the regulation of the cell cycle, we expressed an exogenous CCT2-FLAG construct in T47D and MCF7 luminal A breast cancer cells and examined cell proliferation under conditions of two-dimensional (2D) monolayer and three-dimensional (3D) spheroid cultures. Exogenous CCT2 increased the proliferation of cancer cells, resulting in larger and multiple spheroids as compared to control cells. CCT2-expressing cells were also able to undergo spheroid growth reversal, re-attaching, and resuming growth in 2D cultures. Such cells gained anchorage-independent growth. CCT2 expression in cells correlated with increased expression of MYC, especially in spheroid cultures, and other cell cycle regulators like CCND1 and CDK2, indicative of a novel activity that could contribute to the increase in cell growth. Statistically significant correlations between CCT2, MYC, and CCND1 were shown. Since CCT2 is located on chromosome 12q15, an amplicon frequently found in soft tissue cancers as well as breast cancer, CCT2 may have the basic characteristics of an oncogene. Our findings suggest that CCT2 could be an essential driver of cell division that may be a node through which pathways involving MYC, cyclin D1 and other proliferative factors could converge. Hence the therapeutic inhibition of CCT2 may have the potential to achieve multi-target inhibition, overcoming the limitations associated with single agent inhibitors.
Collapse
Affiliation(s)
- Heba Ghozlan
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Adrian Showalter
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Eunkyung Lee
- Department of Health Sciences, College of Health Professions and Sciences, University of Central Florida, Orlando, FL, United States
| | - Xiang Zhu
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Annette R Khaled
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
4
|
Chong ZX, Yeap SK, Ho WY. Dysregulation of miR-638 in the progression of cancers. Pathol Res Pract 2021; 220:153351. [PMID: 33642053 DOI: 10.1016/j.prp.2021.153351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/14/2022]
Abstract
MicroRNA (miRNA) is a form of short, single-stranded and non-coding RNA that is important in regulating the post-transcriptional modification of multiple downstream targets. Many miRNAs have been reported to involve in controlling the progression of human diseases, and one of them is miR-638, which play essential roles in regulating the development of human cancer. By targeting the 3'-ends of its targets, miR-638 can regulate cellular processes including proliferation, invasion, metastases, angiogenesis, apoptosis and inflammation. This review was aimed to summarize current findings on the roles of miR-638 in different human cancers based on the results from various in vitro, in vivo and clinical studies. The biogenesis process and tissue expression, followed by the roles of miR-638 in regulating the development of various human cancers by targeting different downstream targets were covered in this review. The potential applications and challenges of employing miR-638 as cancer biomarker and therapeutic agent were also discussed.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900, Sepang, Selangor, Malaysia.
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|
5
|
Lim WC, Choi HK, Kim KT, Lim TG. Rose ( Rosa gallica) Petal Extract Suppress Proliferation, Migration, and Invasion of Human Lung Adenocarcinoma A549 Cells through via the EGFR Signaling Pathway. Molecules 2020; 25:molecules25215119. [PMID: 33158043 PMCID: PMC7663240 DOI: 10.3390/molecules25215119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
We sought to investigate the effect of rose petal extract (RPE) on the proliferation, migration, and invasion of cancer cells. RPE significantly inhibited the growth of lung and colorectal cancer cell lines, with rapid suppression of A549 lung cancer cells at low concentrations. These effects occurred concomitantly with downregulation of the cell proliferation mediators PCNA, cyclin D1, and c-myc. In addition, RPE suppressed the migration and invasion of A549 cells by inhibiting the expression and activity of matrix metalloproteinase-2 and matrix metalloproteinase-9 (MMP-2 and -9). We hypothesize that the suppressive activity of RPE against lung cancer cell proliferation and early metastasis occurs via the EGFR-MAPK and mTOR-Akt signaling pathways. These early results highlight the significant potency of RPE, particularly for lung cancer cells, and warrant further investigation.
Collapse
Affiliation(s)
- Won-Chul Lim
- Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Korea; (W.-C.L.); (H.-K.C.); (K.-T.K.)
| | - Hyo-Kyung Choi
- Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Korea; (W.-C.L.); (H.-K.C.); (K.-T.K.)
| | - Kyung-Tack Kim
- Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Korea; (W.-C.L.); (H.-K.C.); (K.-T.K.)
| | - Tae-Gyu Lim
- Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Korea; (W.-C.L.); (H.-K.C.); (K.-T.K.)
- Department of Food Science & Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Korea
- Correspondence: ; Tel.: +82-2-3208-3460
| |
Collapse
|
6
|
Ishikawa K, Kobayashi Y, Wakabayashi Y, Watanabe S, Semba K. A highly sensitive trap vector system for isolating reporter cells and identification of responsive genes. Biol Methods Protoc 2018; 3:bpy003. [PMID: 32161797 PMCID: PMC6994077 DOI: 10.1093/biomethods/bpy003] [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: 02/06/2018] [Revised: 03/22/2018] [Accepted: 04/18/2018] [Indexed: 01/03/2023] Open
Abstract
We devised a versatile vector system for efficient isolation of reporter cells responding to a certain condition of interest. This system combines nontoxic GAL4-UAS and piggyBac transposon systems, allowing application to mammalian cells and improved expression of a fluorescent reporter protein for cell sorting. Case studies under conditions of c-MYC gene induction or endoplasmic reticulum (ER) stress with thapsigargin on mouse or human cell lines confirmed easy and efficient isolation of responsive reporter cells. Sequence analyses of the integrated loci of the thapsigargin-responsive clones identified responsive genes including BiP and OSBPL9. OSBPL9 is a novel ER stress-responsive gene and we confirmed that endogenous mRNA expression of OSBPL9 is upregulated by thapsigargin, and is repressed by IRE1α inhibitors, 4μ8C and toyocamycin, but not significantly by a PERK inhibitor, GSK2656157. These results demonstrate that this approach can be used to discover novel genes regulated by any stimuli without the need for microarray analysis, and that it can concomitantly produce reporter cells without identification of stimuli-responsive promoter/enhancer elements. Therefore, this system has a variety of benefits for basic and clinical research.
Collapse
Affiliation(s)
- Kosuke Ishikawa
- Japan Biological Informatics Consortium (JBiC), 2-45 Aomi, Koto-ku, Tokyo 135-8073, Japan
| | - Yuta Kobayashi
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Yutaro Wakabayashi
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Shinya Watanabe
- Translational Research Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan.,Translational Research Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| |
Collapse
|
7
|
Novel phyto-derivative BRM270 inhibits hepatocellular carcinoma cells proliferation by inducing G2/M phase cell cycle arrest and apoptosis in xenograft mice model. Biomed Pharmacother 2017; 87:741-754. [DOI: 10.1016/j.biopha.2017.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/19/2016] [Accepted: 01/01/2017] [Indexed: 01/06/2023] Open
|
8
|
Liu R, Liu T, Wei W, Guo K, Yang N, Tian S, Zhu J, Liu Y, Zhou W, Yang H. Novel interacting proteins identified by tandem affinity purification coupled to nano LC-MS/MS interact with ribosomal S6 protein kinase 4 (RSK4) and its variant protein (RSK4m). Int J Biol Macromol 2017; 96:421-428. [PMID: 27986634 DOI: 10.1016/j.ijbiomac.2016.12.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 12/16/2022]
Abstract
Ribosomal S6 protein kinase 4 (RSK4) is an important novel tumor suppressor that inhibits breast cancer cell growth and induces senescence. However, RSK4m, which is a variant of RSK4 resulting from alternative splicing, may play distinct roles in some aspects. Knowledge about the mechanisms of RSK4 or RSK4m activity has been lacking. Analysis of the RSK4 and RSK4m interactome could provide insight into their specific functions and integrative mechanisms. Using tandem affinity purification, we obtained protein complexes that interacted with RSK4 or RSK4m. Mass spectrum analysis was performed to identify the obtained protein complexes, and bioinformatics analysis was performed. In this study, we isolated and identified 82 RSK4-associated proteins and 137 RSK4m-associated proteins using two STREP II and a single Flag tag-based tandem affinity purification (SF-TAP) coupled with nano LC-MS/MS in MDA-MB-231 cells. Gene Ontology and Ingenuity Pathway Analysis analyses provided functional annotations and protein-protein interaction networks of the protein interactome of RSK4 and RSK4m. Functional annotations of these proteins by bioinformatics analyses highlight the essential role of RSK4 and RSK4m in coordination with their interacting partners to mediate multiple biological processes, especially cell senescence. Moreover, after comparing the interactome of RSK4 and RSK4m, we found that RSK4m is involved in more molecular functions than RSK4.
Collapse
Affiliation(s)
- Riqiang Liu
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Tianhua Liu
- Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai 200032, China.
| | - Wei Wei
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Kun Guo
- Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Ning Yang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Siqi Tian
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jia Zhu
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yinkun Liu
- Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Wenting Zhou
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Huawei Yang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
9
|
Hu Y, Yang L, Yang Y, Han Y, Wang Y, Liu W, Zuo J. Oncogenic role of mortalin contributes to ovarian tumorigenesis by activating the MAPK-ERK pathway. J Cell Mol Med 2016; 20:2111-2121. [PMID: 27374312 PMCID: PMC5082394 DOI: 10.1111/jcmm.12905] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/07/2016] [Indexed: 12/21/2022] Open
Abstract
Mortalin is frequently overexpressed in human malignancies. Previous studies have suggested that mortalin contributes to ovarian cancer development and progression, but further investigation is warranted. The aim of this study is to elucidate the mechanism of mortalin in ovarian cancer development and progression. In this study, lentivirus‐delivered mortalin short hairpin RNA (shRNA) was used to knockdown mortalin expression in A2780 and A2780/cis ovarian cancer cell lines, and lentiviral mortalin‐pLVX‐AcGFP was used to generate mortalin‐overexpressing cell lines. The results demonstrated that decreased mortalin expression reduced ovarian cancer cell proliferation, colony formation, migration and invasion by Cell Counting Kit‐8 assay, colony formation assay, wounding healing assay and Transwell cell invasion assay, respectively. Flow cytometry results suggested that mortalin promotes the G1 transition, leading to faster restoration of a normal cell‐cycle distribution. Cell‐cycle proteins, including C‐myc and Cyclin‐D1, significantly increased, and Cyclin‐B1 remarkably decreased upon mortalin down‐regulation. Western blot analysis showed that mortalin knockdown significantly decreased p‐c‐Raf and phospho‐extracellular–regulated protein kinases (p‐ERK1/2) pathways but not the Jun N‐terminal kinase pathway, whereas mortalin overexpression had the opposite effect. Taken together, these results indicate that mortalin is an oncogenic factor, and mitogen‐activated protein kinase‐ERK signalling pathway activation by mortalin may contribute to ovarian cancer development and progression.
Collapse
Affiliation(s)
- Yingying Hu
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ling Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yujie Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yanyan Han
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongbo Wang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wen Liu
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Ji Zuo
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Wang F, Remke M, Bhat K, Wong ET, Zhou S, Ramaswamy V, Dubuc A, Fonkem E, Salem S, Zhang H, Hsieh TC, O'Rourke ST, Wu L, Li DW, Hawkins C, Kohane IS, Wu JM, Wu M, Taylor MD, Wu E. A microRNA-1280/JAG2 network comprises a novel biological target in high-risk medulloblastoma. Oncotarget 2015; 6:2709-24. [PMID: 25576913 PMCID: PMC4413612 DOI: 10.18632/oncotarget.2779] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 11/19/2014] [Indexed: 01/23/2023] Open
Abstract
Over-expression of PDGF receptors (PDGFRs) has been previously implicated in high-risk medulloblastoma (MB) pathogenesis. However, the exact biological functions of PDGFRα and PDGFRβ signaling in MB biology remain poorly understood. Here, we report the subgroup specific expression of PDGFRα and PDGFRβ and their associated biological pathways in MB tumors. c-MYC, a downstream target of PDGFRβ but not PDGFRα, is involved in PDGFRβ signaling associated with cell proliferation, cell death, and invasion. Concurrent inhibition of PDGFRβ and c-MYC blocks MB cell proliferation and migration synergistically. Integrated analysis of miRNA and miRNA targets regulated by both PDGFRβ and c-MYC reveals that increased expression of JAG2, a target of miR-1280, is associated with high metastatic dissemination at diagnosis and a poor outcome in MB patients. Our study may resolve the controversy on the role of PDGFRs in MB and unveils JAG2 as a key downstream effector of a PDGFRβ-driven signaling cascade and a potential therapeutic target.
Collapse
Affiliation(s)
- Fengfei Wang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Marc Remke
- Arthur and Sonia Labatt Brain Tumor Research Centre, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Kruttika Bhat
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Eric T Wong
- Brain Tumor Center & Neuro-Oncology Unit, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Shuang Zhou
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Vijay Ramaswamy
- Arthur and Sonia Labatt Brain Tumor Research Centre, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Adrian Dubuc
- Arthur and Sonia Labatt Brain Tumor Research Centre, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Ekokobe Fonkem
- Scott & White Neuroscience Institute, Texas A & M Health Science Center, Temple, TX 76508, USA
| | - Saeed Salem
- Department of Computer Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Hongbing Zhang
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100073, China
| | - Tze-Chen Hsieh
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Stephen T O'Rourke
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, Shands Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - David W Li
- Department of Ophthalmology & Visual Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Cynthia Hawkins
- Division of Pathology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Isaac S Kohane
- Informatics Program, Children's Hospital Boston, Harvard Medical School, Boston 02115, MA, USA
| | - Joseph M Wu
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Min Wu
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, ND 58202, USA
| | - Michael D Taylor
- Arthur and Sonia Labatt Brain Tumor Research Centre, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
11
|
Kourtidis A, Ngok SP, Pulimeno P, Feathers RW, Carpio LR, Baker TR, Carr JM, Yan IK, Borges S, Perez EA, Storz P, Copland JA, Patel T, Thompson EA, Citi S, Anastasiadis PZ. Distinct E-cadherin-based complexes regulate cell behaviour through miRNA processing or Src and p120 catenin activity. Nat Cell Biol 2015; 17:1145-57. [PMID: 26302406 PMCID: PMC4975377 DOI: 10.1038/ncb3227] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 07/20/2015] [Indexed: 12/11/2022]
Abstract
E-cadherin and p120 catenin (p120) are essential for epithelial homeostasis, but can also exert pro-tumorigenic activities. Here, we resolve this apparent paradox by identifying two spatially and functionally distinct junctional complexes in non-transformed polarized epithelial cells: one growth suppressing at the apical zonula adherens (ZA), defined by the p120 partner PLEKHA7 and a non-nuclear subset of the core microprocessor components DROSHA and DGCR8, and one growth promoting at basolateral areas of cell-cell contact containing tyrosine-phosphorylated p120 and active Src. Recruitment of DROSHA and DGCR8 to the ZA is PLEKHA7 dependent. The PLEKHA7-microprocessor complex co-precipitates with primary microRNAs (pri-miRNAs) and possesses pri-miRNA processing activity. PLEKHA7 regulates the levels of select miRNAs, in particular processing of miR-30b, to suppress expression of cell transforming markers promoted by the basolateral complex, including SNAI1, MYC and CCND1. Our work identifies a mechanism through which adhesion complexes regulate cellular behaviour and reveals their surprising association with the microprocessor.
Collapse
Affiliation(s)
- Antonis Kourtidis
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Siu P. Ngok
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Pamela Pulimeno
- Department of Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, CH-1211, Geneva 4, Switzerland
| | - Ryan W. Feathers
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Lomeli R. Carpio
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Tiffany R. Baker
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Jennifer M. Carr
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Irene K. Yan
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Sahra Borges
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Edith A. Perez
- Division of Hematology/Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Tushar Patel
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - E. Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Sandra Citi
- Department of Cell Biology and Institute of Genetics and Genomics of Geneva, University of Geneva, 30 quai Ernest-Ansermet, CH-1211, Geneva 4, Switzerland
| | - Panos Z. Anastasiadis
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| |
Collapse
|
12
|
Schueler M, Munschauer M, Gregersen LH, Finzel A, Loewer A, Chen W, Landthaler M, Dieterich C. Differential protein occupancy profiling of the mRNA transcriptome. Genome Biol 2014; 15:R15. [PMID: 24417896 PMCID: PMC4056462 DOI: 10.1186/gb-2014-15-1-r15] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 01/13/2014] [Indexed: 12/16/2022] Open
Abstract
Background RNA-binding proteins (RBPs) mediate mRNA biogenesis, translation and decay. We recently developed an approach to profile transcriptome-wide RBP contacts on polyadenylated transcripts by next-generation sequencing. A comparison of such profiles from different biological conditions has the power to unravel dynamic changes in protein-contacted cis-regulatory mRNA regions without a priori knowledge of the regulatory protein component. Results We compared protein occupancy profiles of polyadenylated transcripts in MCF7 and HEK293 cells. Briefly, we developed a bioinformatics workflow to identify differential crosslinking sites in cDNA reads of 4-thiouridine crosslinked polyadenylated RNA samples. We identified 30,000 differential crosslinking sites between MCF7 and HEK293 cells at an estimated false discovery rate of 10%. 73% of all reported differential protein-RNA contact sites cannot be explained by local changes in exon usage as indicated by complementary RNA-seq data. The majority of differentially crosslinked positions are located in 3′ UTRs, show distinct secondary-structure characteristics and overlap with binding sites of known RBPs, such as ELAVL1. Importantly, mRNA transcripts with the most significant occupancy changes show elongated mRNA half-lives in MCF7 cells. Conclusions We present a global comparison of protein occupancy profiles from different cell types, and provide evidence for altered mRNA metabolism as a result of differential protein-RNA contacts. Additionally, we introduce POPPI, a bioinformatics workflow for the analysis of protein occupancy profiling experiments. Our work demonstrates the value of protein occupancy profiling for assessing cis-regulatory RNA sequence space and its dynamics in growth, development and disease.
Collapse
|
13
|
Sun Y, Lou X, Yang M, Yuan C, Ma L, Xie BK, Wu JM, Yang W, Shen SX, Xu N, Liao DJ. Cyclin-dependent kinase 4 may be expressed as multiple proteins and have functions that are independent of binding to CCND and RB and occur at the S and G 2/M phases of the cell cycle. Cell Cycle 2013; 12:3512-25. [PMID: 24091631 DOI: 10.4161/cc.26510] [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
Cyclin-dependent kinase 4 (CDK4) is known to be a 33 kD protein that drives G 1 phase progression of the cell cycle by binding to a CCND protein to phosphorylate RB proteins. Using different CDK4 antibodies in western blot, we detected 2 groups of proteins around 40 and 33 kD, respectively, in human and mouse cells; each group often appeared as a duplet or triplet of bands. Some CDK4 shRNAs could decrease the 33 kD wild-type (wt) CDK4 but increase some 40 kD proteins, whereas some other shRNAs had the opposite effects. Liquid chromatography-mass spectrometry/mass spectrometry analysis confirmed the existence of CDK4 isoforms smaller than 33 kD but failed to identify CDK4 at 40 kD. We cloned one CDK4 mRNA variant that lacks exon 2 and encodes a 26 kD protein without the first 74 amino acids of the wt CDK4, thus lacking the ATP binding sequence and the PISTVRE domain required for binding to CCND. Co-IP assay confirmed that this ΔE2 protein lost CCND1- and RB1-binding ability. Moreover, we found, surprisingly, that the wt CDK4 and the ΔE2 could inhibit G 1-S progression, accelerate S-G 2/M progression, and enhance or delay apoptosis in a cell line-specific manner in a situation where the cells were treated with a CDK4 inhibitor or the cells were serum-starved and then replenished. Hence, CDK4 seems to be expressed as multiple proteins that react differently to different CDK4 antibodies, respond differently to different shRNAs, and, in some situations, have previously unrecognized functions at the S-G 2/M phases of the cell cycle via mechanisms independent of binding to CCND and RB.
Collapse
Affiliation(s)
- Yuan Sun
- Hormel Institute; The University of Minnesota; Austin, MN USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Tehrani GA, Mirzaahmadi S, Bandehpour M, Kazemi B. Coexpression of luxA and luxB genes of Vibrio fischeri in NIH3T3 mammalian cells and evaluation of its bioluminescence activities. LUMINESCENCE 2013; 29:13-9. [PMID: 23616465 DOI: 10.1002/bio.2468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Revised: 10/01/2012] [Accepted: 11/17/2012] [Indexed: 11/05/2022]
Abstract
Expression of bacterial luciferase enzyme (lux) in eukaryotic cells would provide a new bioreporter system for in vivo imaging and diagnostics technology. In spite of this, until now only a few efforts have been made to express bacterial luciferase enzyme in eukaryotic cells. We attempted to synthesize an expression construct of luxA and luxB genes from Vibrio fischeri. The luxA and luxB genes were cloned into the MCS of pTZ57R via the 5' kpnI, BamHI and BamHI, EcoRI restriction sites to generate pTZ57R/luxA and pTZ57R/luxB respectively, then newly synthesized constructs were cleaved with the same enzymes and respectively cloned into the pcDNA3.1(+) (Hyg) and pcDNA3.1(+) (Neo) expression vectors to create pcDNA3.1(+) (Hyg)/luxA and pcDNA3.1(+) (neo)/luxB. Recombinant constructs were cotransfected to the NIH3T3 cell line. Gene expression was confirmed by reverse transcription-polymerase chain reaction, sodium dodecyl sulfate-polyacrylamide gel electrophoresis and Western blotting; in addition, bioluminescence characteristics of transfected NIH3T3 cell lines were evaluated by decanal supplement. In conclusion, in the current research, separate vector systems were constructed, which are composed of bacterial luciferase genes (luxA and luxB) that accordingly have not already been reported. These results hold promise toward the potential development of an autonomous light-generating lux reporter system in eukaryotic cells.
Collapse
|
15
|
Sun Y, Li Y, Luo D, Liao DJ. Pseudogenes as weaknesses of ACTB (Actb) and GAPDH (Gapdh) used as reference genes in reverse transcription and polymerase chain reactions. PLoS One 2012; 7:e41659. [PMID: 22927912 PMCID: PMC3425558 DOI: 10.1371/journal.pone.0041659] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/25/2012] [Indexed: 01/19/2023] Open
Abstract
The genes encoding β-actin (ACTB in human or Actb in mouse) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH in human or Gapdh in mouse) are the two most commonly used references for sample normalization in determination of the mRNA level of interested genes by reverse transcription (RT) and ensuing polymerase chain reactions (PCR). In this study, bioinformatic analyses revealed that the ACTB, Actb, GAPDH and Gapdh had 64, 69, 67 and 197 pseudogenes (PGs), respectively, in the corresponding genome. Most of these PGs are intronless and similar in size to the authentic mRNA. Alignment of several PGs of these genes with the corresponding mRNA reveals that they are highly homologous. In contrast, the hypoxanthine phosphoribosyltransferase-1 gene (HPRT1 in human or Hprt in mouse) only had 3 or 1 PG, respectively, and the mRNA has unique regions for primer design. PCR with cDNA or genomic DNA (gDNA) as templates revealed that our HPRT1, Hprt and GAPDH primers were specific, whereas our ACTB and Actb primers were not specific enough both vertically (within the cDNA) and horizontally (compared cDNA with gDNA). No primers could be designed for the Gapdh that would not mis-prime PGs. Since most of the genome is transcribed, we suggest to peers to forgo ACTB (Actb) and GAPDH (Dapdh) as references in RT-PCR and, if there is no surrogate, to use our primers with extra caution. We also propose a standard operation procedure in which design of primers for RT-PCR starts from avoiding mis-priming PGs and all primers need be tested for specificity with both cDNA and gDNA.
Collapse
Affiliation(s)
- Yuan Sun
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
- Department of Pathology, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Yan Li
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Dianzhong Luo
- Department of Pathology, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
- * E-mail: (DZL); (DJL)
| | - D. Joshua Liao
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
- * E-mail: (DZL); (DJL)
| |
Collapse
|
16
|
Zhang T, Qi Y, Liao M, Xu M, Bower KA, Frank JA, Shen HM, Luo J, Shi X, Chen G. Autophagy Is a Cell Self-Protective Mechanism Against Arsenic-Induced Cell Transformation. Toxicol Sci 2012; 130:298-308. [DOI: 10.1093/toxsci/kfs240] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
17
|
Sun Y, Cao S, Yang M, Wu S, Wang Z, Lin X, Song X, Liao DJ. Basic anatomy and tumor biology of the RPS6KA6 gene that encodes the p90 ribosomal S6 kinase-4. Oncogene 2012; 32:1794-810. [PMID: 22614021 PMCID: PMC3427418 DOI: 10.1038/onc.2012.200] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The RPS6KA6 gene encodes the p90 ribosomal S6 kinase-4 (RSK4) that is still largely uncharacterized. In this study we identified a new RSK4 transcription initiation site and several alternative splice sites with a 5’RACE approach. The resulting mRNA variants encompass four possible first start codons. The first 15 nucleotides (nt) of exon 22 in mouse and the penultimate exon in both human (exon 21) and mouse (exon 24) RSK4 underwent alternative splicing, although the penultimate exon deleted variant appeared mainly in cell clines, but not in most normal tissues. Demethylation agent 5-azacytidine inhibited the deletion of the penultimate exon whereas two indolocarbazole-derived inhibitors of cyclin dependent kinase 4 or 6 induced deletion of the first 39 nt from exon 21 of human RSK4. In all human cancer cell lines studied, the 90-kD wild type RSK4 was sparse but, surprisingly, several isoforms at or smaller than 72-kD were expressed as detected by seven different antibodies. On immunoblots, each of these smaller isoforms often appeared as a duplet or triplet and the levels of these isoforms varied greatly among different cell lines and culture conditions. Cyclin D1 inhibited RSK4 expression and serum starvation enhanced the inhibition, whereas c-Myc and RSK4 inhibited cyclin D1. The effects of RSK4 on cell growth, cell death and chemoresponse depended on the mRNA variant or the protein isoform expressed, on the specificity of the cell lines, as well as on the anchorage-dependent or -independent growth conditions and the in vivo situation. Moreover, we also observed that even a given cDNA might be expressed to multiple proteins; therefore, when using a cDNA, one needs to exclude this possibility before attribution of the biological results from the cDNA to the anticipated protein. Collectively, our results suggest that whether RSK4 is oncogenic or tumor suppressive depends on many factors.
Collapse
Affiliation(s)
- Y Sun
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Fung FKC, Chan DW, Liu VWS, Leung THY, Cheung ANY, Ngan HYS. Increased expression of PITX2 transcription factor contributes to ovarian cancer progression. PLoS One 2012; 7:e37076. [PMID: 22615897 PMCID: PMC3352869 DOI: 10.1371/journal.pone.0037076] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 04/13/2012] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Paired-like homeodomain 2 (PITX2) is a bicoid homeodomain transcription factor which plays an essential role in maintaining embryonic left-right asymmetry during vertebrate embryogenesis. However, emerging evidence suggests that the aberrant upregulation of PITX2 may be associated with tumor progression, yet the functional role that PITX2 plays in tumorigenesis remains unknown. PRINCIPAL FINDINGS Using real-time quantitative RT-PCR (Q-PCR), Western blot and immunohistochemical (IHC) analyses, we demonstrated that PITX2 was frequently overexpressed in ovarian cancer samples and cell lines. Clinicopathological correlation showed that the upregulated PITX2 was significantly associated with high-grade (P = 0.023) and clear cell subtype (P = 0.011) using Q-PCR and high-grade (P<0.001) ovarian cancer by IHC analysis. Functionally, enforced expression of PITX2 could promote ovarian cancer cell proliferation, anchorage-independent growth ability, migration/invasion and tumor growth in xenograft model mice. Moreover, enforced expression of PITX2 elevated the cell cycle regulatory proteins such as Cyclin-D1 and C-myc. Conversely, RNAi mediated knockdown of PITX2 in PITX2-high expressing ovarian cancer cells had the opposite effect. CONCLUSION Our findings suggest that the increased expression PITX2 is involved in ovarian cancer progression through promoting cell growth and cell migration/invasion. Thus, targeting PITX2 may serve as a potential therapeutic modality in the management of high-grade ovarian tumor.
Collapse
Affiliation(s)
- Frederic K. C. Fung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - David W. Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Vincent W. S. Liu
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Thomas H. Y. Leung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Annie N. Y. Cheung
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Hextan Y. S. Ngan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| |
Collapse
|
19
|
Galatola M, Paparo L, Duraturo F, Turano M, Rossi GB, Izzo P, De Rosa M. Beta catenin and cytokine pathway dysregulation in patients with manifestations of the "PTEN hamartoma tumor syndrome". BMC MEDICAL GENETICS 2012; 13:28. [PMID: 22520842 PMCID: PMC3353236 DOI: 10.1186/1471-2350-13-28] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 04/20/2012] [Indexed: 12/24/2022]
Abstract
Background The "PTEN hamartoma tumor syndrome" (PHTS) includes a group of syndromes caused by germline mutations within the tumor suppressor gene "phosphatase and tensin homolog deleted on chromosome ten" (PTEN), characterized by multiple polyps in the gastrointestinal tract and by a highly increased risk of developing malignant tumours in many tissues. The current work clarifies the molecular basis of PHTS in three unrelated Italian patients, and sheds light on molecular pathway disregulation constitutively associated to PTEN alteration. Methods We performed a combination of RT-PCR, PCR, sequencing of the amplified fragments, Real Time PCR and western blot techniques. Results Our data provide the first evidence of β-catenin accumulation in blood cells of patients with hereditary cancer syndrome caused by germ-line PTEN alteration. In addition, for the first time we show, in all PHTS patients analysed, alterations in the expression of TNFα, its receptors and IL-10. Importantly, the isoform of TNFRI that lacks the DEATH domain (TNFRSF1β) was found to be overexpressed. Conclusion In light of our findings, we suggest that the PTEN pathway disregulation could determine, in non-neoplastic cells of PHTS patients, cell survival and pro-inflammatory stimulation, mediated by the expression of molecules such as β-catenin, TNFα and TNFα receptors, which could predispose these patients to the development of multiple cancers.
Collapse
Affiliation(s)
- Martina Galatola
- Dipartimento di Biochimica e Biotecnologie Mediche and CEINGE Biotecnologie Avanzate, Università di Napoli Federico II, via S. Pansini 5, Naples 80131, Italy
| | | | | | | | | | | | | |
Collapse
|
20
|
Drew BA, Burow ME, Beckman BS. MEK5/ERK5 pathway: the first fifteen years. Biochim Biophys Acta Rev Cancer 2011; 1825:37-48. [PMID: 22020294 DOI: 10.1016/j.bbcan.2011.10.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 10/07/2011] [Indexed: 12/22/2022]
Abstract
While conventional MAP kinase pathways are one of the most highly studied signal transduction molecules, less is known about the MEK5 signaling pathway. This pathway has been shown to play a role in normal cell growth cycles, survival and differentiation. The MEK5 pathway is also believed to mediate the effects of a number of oncogenes. MEK5 is the upstream activator of ERK5 in many epithelial cells. Activation of the MEK-MAPK pathway is a frequent event in malignant tumor formation and contributes to chemoresistance and anti-apoptotic signaling. This pathway may be involved in a number of more aggressive, metastatic varieties of cancer due to its role in cell survival, proliferation and EMT transitioning. Further study of this pathway may lead to new prognostic factors and new drug targets to combat more aggressive forms of cancer.
Collapse
Affiliation(s)
- Barbara A Drew
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
21
|
Yang M, Sun Y, Ma L, Wang C, Wu JM, Bi A, Liao DJ. Complex alternative splicing of the smarca2 gene suggests the importance of smarca2-B variants. J Cancer 2011; 2:386-400. [PMID: 21811517 PMCID: PMC3148773 DOI: 10.7150/jca.2.386] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/22/2011] [Indexed: 11/21/2022] Open
Abstract
BRM is an ATPase component of the SWI/SNF complex that regulates chromatin remodeling and cell proliferation and is considered a tumor suppressor. In this study we characterized transcripts from the Smarca2 gene that encodes the BRM protein. We found that the human Smarca2 gene (hSmarca2), like its mouse counterpart (mSmarca2), also initiated a short transcript from intron 27 of the long transcript. We name the long and short transcripts as Smarca2-a and Smarca2-b, respectively. Like its human counterpart, mSmarca2-a also underwent alternative splicing at the 54-bp exon 29. The hSmarca2-b had two alternative initiation sites and underwent alternative splicing at three different 3' sites of exon 1 and at exons 2, 3 and/or 5. We identified nine hSmarca2-b mRNA variants that might produce five different proteins. mSmarca2-b also underwent alternative splicing at exon 3 and/or exon 5, besides alternatively retaining part of intron 1 in exon 1. Smarca2-b was expressed more abundantly than Smarca2-a in many cell lines and was more sensitive to serum starvation. Moreover, cyclin D1 also regulated the expression of both Smarca2-a and Smarca2-b in a complex manner. These data suggest that the functions of the Smarca2 gene may be very complex, not just simply inhibiting cell proliferation, and in certain situations may be elicited mainly by expressing the much less known Smarca2-b, not the better studied Smarca2-a and its products BRM proteins.
Collapse
Affiliation(s)
- Min Yang
- 1. Hormel Institute, The University of Minnesota, Austin, MN 55912, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Dombkowski AA, Sultana Z, Craig DB, Jamil H. In silico analysis of combinatorial microRNA activity reveals target genes and pathways associated with breast cancer metastasis. Cancer Inform 2011; 10:13-29. [PMID: 21552493 PMCID: PMC3085424 DOI: 10.4137/cin.s6631] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
This is an open access article. Unrestricted non-commercial use is permitted provided the original work is properly cited. Aberrant microRNA activity has been reported in many diseases, and studies often find numerous microRNAs concurrently dysregulated. Most target genes have binding sites for multiple microRNAs, and mounting evidence indicates that it is important to consider their combinatorial effect on target gene repression. A recent study associated the coincident loss of expression of six microRNAs with metastatic potential in breast cancer. Here, we used a new computational method, miR-AT!, to investigate combinatorial activity among this group of microRNAs. We found that the set of transcripts having multiple target sites for these microRNAs was significantly enriched with genes involved in cellular processes commonly perturbed in metastatic tumors: cell cycle regulation, cytoskeleton organization, and cell adhesion. Network analysis revealed numerous target genes upstream of cyclin D1 and c-Myc, indicating that the collective loss of the six microRNAs may have a focal effect on these two key regulatory nodes. A number of genes previously implicated in cancer metastasis are among the predicted combinatorial targets, including TGFB1, ARPC3, and RANKL. In summary, our analysis reveals extensive combinatorial interactions that have notable implications for their potential role in breast cancer metastasis and in therapeutic development.
Collapse
Affiliation(s)
- Alan A Dombkowski
- Division of Clinical Pharmacology and Toxicology, Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
| | | | | | | |
Collapse
|
23
|
Wang C, Lisanti MP, Liao DJ. Reviewing once more the c-myc and Ras collaboration: converging at the cyclin D1-CDK4 complex and challenging basic concepts of cancer biology. Cell Cycle 2011; 10:57-67. [PMID: 21200143 DOI: 10.4161/cc.10.1.14449] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The c-myc is a proto-oncogene that manifests aberrant expression at high frequencies in most types of human cancer. C-myc gene amplifications are often observed in various cancers as well. Ample studies have also proved that c-myc has a potent oncogenicity, which can be further enhanced by collaborations with other oncogenes such as Bcl-2 and activated Ras. Studies on the collaborations of c-myc with Ras or other genes in oncogenicity have established several basic concepts and have disclosed their underlying mechanisms of tumor biology, including "immortalization" and "transformation". In many cases, these collaborations may converge at the cyclin D1-CDK4 complex. In the meantime, however, many results from studies on the c-myc, Ras and cyclin D1-CDK4 also challenge these basic concepts of tumor biology and suggest to us that the immortalized status of cells should be emphasized. Stricter criteria and definitions for a malignantly transformed status and a benign status of cells in culture also need to be established to facilitate our study of the mechanisms for tumor formation and to better link up in vitro data with animal results and eventually with human cancer pathology.
Collapse
Affiliation(s)
- Chenguang Wang
- Department of Stem Cell and Regenerative Medicine, and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | |
Collapse
|
24
|
Cho KB, Cho MK, Lee WY, Kang KW. Overexpression of c-myc induces epithelial mesenchymal transition in mammary epithelial cells. Cancer Lett 2010; 293:230-9. [PMID: 20144848 DOI: 10.1016/j.canlet.2010.01.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 01/15/2010] [Accepted: 01/16/2010] [Indexed: 01/15/2023]
Abstract
The c-myc gene is frequently overexpressed in human breast cancer and its target genes are involved in tumorigenesis. Epithelial mesenchymal transitions (EMT), where cells undergo a developmental switch from a polarized epithelial phenotype to a highly motile mesenchymal phenotype, are associated with invasion and motility of cancer cells. Basal E-cadherin expression was down-regulated in c-myc overexpressing MCF10A (c-myc-MCF10A) cells compared to GFP-overexpressing MCF10A (GFP-MCF10A) cells, while N-cadherin was distinctly increased in c-myc-MCF10A cells. Given that glycogen synthase kinase-3beta (GSK-3beta) and the snail axis have key roles in E-cadherin deregulation during EMT, we investigated the role of GSK-3beta/snail signaling pathways in the induction of EMT by c-myc overexpression. In contrast to GFP-MCF10A cells, both the transcriptional activity and the ubiquitination-dependent protein stability of snail were enhanced in c-myc-MCF10A cells, and this was reversed by GSK-3beta overexpression. We also found that c-myc overexpression inhibits GSK-3beta activity through activation of extracellular signal-regulated kinase (ERK). Inhibition of ERK by dominant negative mutant transfection or chemical inhibitor significantly suppressed snail gene transcription. These results suggest that c-myc overexpression during transformation of mammary epithelial cells (MEC) is involved in EMTs via ERK-dependent GSK-3beta inactivation and subsequent snail activation.
Collapse
Affiliation(s)
- Kyoung Bin Cho
- BK21 Project Team, College of Pharmacy, Chosun University, Seosuk-dong, Dong-gu, Gwangju 501-759, Republic of Korea
| | | | | | | |
Collapse
|
25
|
Wang Y, Wang X, Liu L, Wang X. HDAC inhibitor trichostatin A-inhibited survival of dopaminergic neuronal cells. Neurosci Lett 2009; 467:212-6. [PMID: 19835929 DOI: 10.1016/j.neulet.2009.10.037] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 09/30/2009] [Accepted: 10/09/2009] [Indexed: 11/28/2022]
Abstract
Histone deacetylase (HDAC) inhibitors have been shown associated with neurodegenerative diseases. However, their effects on survival of dopaminergic neurons remain uncertain. In the present study, the HDAC inhibitor trichostatin A (TSA) was tested in following dopaminergic neuronal cell lines: rat N27, mouse MN9D, and human SH-SY5Y cells. Results demonstrated that a single TSA treatment resulted in decreased cell survival and increased apoptosis in dopaminergic neuronal cells. Pre-treatment with TSA resulted in exacerbated neurotoxic damage to dopaminergic neurons induced by 1-methyl-4-phenylpyridinium and rotenone. These results suggest that HDAC inhibitors may influence Parkinson's disease pathogenesis by inhibiting survival and increasing vulnerability of dopaminergic neurons to neurotoxins. Our data also suggested the importance of prudent use of HDAC inhibitors in therapy.
Collapse
Affiliation(s)
- Yong Wang
- Department of Physiology and Key Laboratory of the Neurodegenerative Disorders of the Chinese Ministry of Education, Capital Medical University, Youanmen, Beijing 100069, China
| | | | | | | |
Collapse
|
26
|
Abstract
Both prolactin (PRL) and estrogen (E2) are involved in the pathogenesis and progression of mammary neoplasia, but the mechanisms by which these hormones interact to exert their effects in breast cancer cells are not well understood. We show here that PRL is able to activate the unliganded estrogen receptor (ER). In breast cancer cells, PRL activates a reporter plasmid containing estrogen response elements (EREs) and induces the ER target gene pS2. These actions are blocked by the antagonist ICI 182,780, showing that ER is required for the PRL-mediated effect. Moreover, PRL leads to phosphorylation of ERalpha in serine-118 (P-ERalpha), a modification related to the potentiation of ligand-independent transcriptional activation. In addition, PRL mimics the effect of E2 on target gene expression by inducing cyclical recruitment of ERalpha and P-ERalpha to ERE-containing promoters, resulting in recruitment of co-activators and acetylation of histone H3. Finally, PRL induces expression of c-Myc and Cyclin D1 and leads to increased cell proliferation, which is specifically antagonized by ICI 182,780 or ERalpha depletion. These results show that ligand-independent ERalpha activation appears to be an important component of the proliferative and transcriptional actions of PRL in breast cancer cells.
Collapse
|
27
|
Identification of the cyclin D1b mRNA variant in mouse. Mol Biol Rep 2008; 36:953-7. [PMID: 18446443 DOI: 10.1007/s11033-008-9267-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Accepted: 04/22/2008] [Indexed: 10/22/2022]
Abstract
Cyclin D1 plays a key regulatory role during the G1 phase of the cell cycle and its gene is amplified and over-expressed in many cancers. The cyclin D1b mRNA variant was established in human cells and recent functional analyses revealed that its protein product harbors unique activities in human cancer cells. By performing reverse transcription-polymerase chain reaction (RT-PCR) and rapid amplification of cDNA ends (RACE) experiments, we identified the cyclin D1b mRNA variant in mouse. Similar to its human counterpart, the mouse cyclin D1b transcript consists of exon 1, 2, 3, 4 and part of intron 4, and contains a long open reading frame (ORF). The predicted peptide from this ORF is 34-amino acid longer than the human cyclin D1b. The expression of this mouse mRNA variant was investigated. It appears to be expressed ubiquitously and differentially in various mouse cell lines and tissues and its level might be proportional to that of the canonical endogenous cyclin D1a mRNA.
Collapse
|