1
|
Guo S, Ding R, Zhao Q, Wang X, Lv S, Ji XY. Recent Insights into the Roles of PEST-Containing Nuclear Protein. Mol Biotechnol 2025; 67:1800-1813. [PMID: 38762838 DOI: 10.1007/s12033-024-01188-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
PEST-containing nuclear protein (PCNP), a short-lived small nuclear protein with 178 amino acids, is a nuclear protein containing two PEST sequences. PCNP is highly expressed in several malignant tumors such as cervical cancer, rectal cancer, and lung cancer. It is also associated with cell cycle regulation and the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and Wnt signaling pathways during tumor growth. The present article discuss how PCNP regulates the PI3K/AKT/mTOR and Wnt signaling pathways and related proteins, and the ubiquitination of PCNP regulates tumor cell cycle as well as the progress of the application of PCNP in the pathophysiology and treatment of colon cancer, human ovarian cancer, thyroid cancer, lung adenocarcinoma and oral squamous cell carcinoma. The main relevant articles were retrieved from PubMed, with keywords such as PEST-containing nuclear protein (PCNP), cancer (tumor), and signaling pathways as inclusion/exclusion criteria. Relevant references has been included and cited in the manuscript.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Ruidong Ding
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Qian Zhao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Xu Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Shuangyu Lv
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
- Kaifeng Key Laboratory for Infectious Diseases and Biosafety, Kaifeng, 475004, Henan, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Mazhai, Erqi District, Zhengzhou, 450064, Henan, China.
| |
Collapse
|
2
|
Scharr M, Hirt B, Neckel PH. Spatial gene expression profile of Wnt-signaling components in the murine enteric nervous system. Front Immunol 2024; 15:1302488. [PMID: 38322254 PMCID: PMC10846065 DOI: 10.3389/fimmu.2024.1302488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
Introduction Wnt-signaling is a key regulator of stem cell homeostasis, extensively studied in the intestinal crypt and other metazoan tissues. Yet, there is hardly any data available on the presence of Wnt-signaling components in the adult enteric nervous system (ENS) in vivo. Methods Therefore, we employed RNAscope HiPlex-assay, a novel and more sensitive in situ hybridization technology. By amplifying target specific signals, this technique enables the detection of low abundance, tightly regulated RNA content as is the case for Wnt-signaling components. Additionally, we compared our data to previously published physiological single cell RNA and RiboTag-based RNA sequencing analyses of enteric gliosis using data-mining approaches. Results Our descriptive analysis shows that several components of the multidi-mensional regulatory network of the Wnt-signaling pathway are present in the murine ENS. The transport and secretion protein for Wnt-ligands Wntless as well as canonical (Wnt3a and Wnt2b) and non-canonical Wnt-ligands (Wnt5a, Wnt7a, Wnt8b and Wnt11) are detectable within submucosal and myenteric plexus. Further, corresponding Frizzled receptors (Fzd1, Fzd3, Fzd6, and Fzd7) and regulatory signaling mediators like R-Spondin/DKK ligands are present in the ENS of the small and large intestine. Further, data mining approaches revealed, that several Wnt-related molecules are expressed by enteric glial cell clusters and are dynamically regulated during the inflammatory manifestation of enteric gliosis. Discussion Our results suggest, that canonical and non-canonical Wnt-signaling has a much broader impact on the mature ENS and its cellular homeostasis in health and inflammation, than previously anticipated.
Collapse
Affiliation(s)
| | | | - Peter H. Neckel
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Tadić V, Zhang W, Brozovic A. The high-grade serous ovarian cancer metastasis and chemoresistance in 3D models. Biochim Biophys Acta Rev Cancer 2024; 1879:189052. [PMID: 38097143 DOI: 10.1016/j.bbcan.2023.189052] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most frequent and aggressive type of epithelial ovarian cancer, with high recurrence rate and chemoresistance being the main issues in its clinical management. HGSOC is specifically challenging due to the metastatic dissemination via spheroids in the ascitic fluid. The HGSOC spheroids represent the invasive and chemoresistant cellular fraction, which is impossible to investigate in conventional two-dimensional (2D) monolayer cell cultures lacking critical cell-to-cell and cell-extracellular matrix interactions. Three-dimensional (3D) HGSOC cultures, where cells aggregate and exhibit relevant interactions, offer a promising in vitro model of peritoneal metastasis and multicellular drug resistance. This review summarizes recent studies of HGSOC in 3D culture conditions and highlights the role of multicellular HGSOC spheroids and ascitic environment in HGSOC metastasis and chemoresistance.
Collapse
Affiliation(s)
- Vanja Tadić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia
| | - Wei Zhang
- Department of Engineering Mechanics, Dalian University of Technology, Linggong Road 2, Dalian CN-116024, China
| | - Anamaria Brozovic
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia.
| |
Collapse
|
4
|
Nano-Strategies Targeting the Integrin αvβ3 Network for Cancer Therapy. Cells 2021; 10:cells10071684. [PMID: 34359854 PMCID: PMC8307885 DOI: 10.3390/cells10071684] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Integrin αvβ3, a cell surface receptor, participates in signaling transduction pathways in cancer cell proliferation and metastasis. Several ligands bind to integrin αvβ3 to regulate proliferation and metastasis in cancer cells. Crosstalk between the integrin and other signal transduction pathways also plays an important role in modulating cancer proliferation. Carcinoembryonic antigen cell adhesion molecule 6 (CEACAM6) activates the downstream integrin FAK to stimulate biological activities including cancer proliferation and metastasis. Blockage of signals related to integrin αvβ3 was shown to be a promising target for cancer therapies. 3,3′,5,5′-tetraiodothyroacetic acid (tetrac) completely binds to the integrin with the thyroid hormone to suppress cancer proliferation. The (E)-stilbene analog, resveratrol, also binds to integrin αvβ3 to inhibit cancer growth. Recently, nanotechnologies have been used in the biomedical field for detection and therapeutic purposes. In the current review, we show and evaluate the potentiation of the nanomaterial carrier RGD peptide, derivatives of PLGA-tetrac (NDAT), and nanoresveratrol targeting integrin αvβ3 in cancer therapies.
Collapse
|
5
|
Chen TJ, Zheng Q, Gao F, Yang T, Ren H, Li Y, Chen MW. MicroRNA-665 facilitates cell proliferation and represses apoptosis through modulating Wnt5a/β-Catenin and Caspase-3 signaling pathways by targeting TRIM8 in LUSC. Cancer Cell Int 2021; 21:215. [PMID: 33858426 PMCID: PMC8051054 DOI: 10.1186/s12935-021-01913-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
Background MicroRNAs (miRNAs) are involved in the oncogenesis, development and transformation of lung squamous cell carcinoma (LUSC). miR-665 is clinically significant and acts as a pivotal function in some cancers. Nevertheless, the effects and the potential mechanisms of miR-665 in human LUSC are still unknown. Methods To analyse the clinical significant of miR-665 in human LUSC, quantitative real-time PCR (qRT-PCR) was use to measure miR-665 expression in LUSC specimen tissues and cell lines. Tripartite motif 8 (TRIM8) was verified a target of miR-665 by performing bioinformatic prediction and luciferase reporter assay. The expression levels of TRIM8 were examined through qRT-PCR and Western blotting in LUSC specimen tissues. CCK8 assay was fulfilled for analyzing the function in LUSC cell proliferation. Flow cytometry was used to detect cell and apoptosis. TRIM8 silencing and overexpression further verified the biological effects as those caused by miR-665. Results Here we reported that miR-665 expression was upregulated in LUSC specimen tissues and cell lines. High miR-665 levels were related to differentiation, tumor size and TNM stage. miR-665 mimics facilitated LUSC cell growth and cell cycle G1-S transition and repressed apoptosis. miR-665 inhibitor suppressed cell proliferation and G1-S transition and promoted apoptosis. miR-665 expression was negatively correlated with TRIM8 mRNA expression in LUSC. Luciferase reporter assay confirmed that TRIM8 was a direct target gene of miR-665. miR-665 mimics downregulated the TRIM8 levels, and miR-665 inhibitor upregulated the TRIM8 levels in LUSC cells. Particularly, silencing TRIM8 led to the similar effects of miR-665 mimics in LUSC cells. Overexpression of TRIM8 inhibited LUSC cell proliferation in vitro and in vivo. Furthermore, miR-665 promoted LUSC cell proliferation through facilitating the Wnt5a/β-catenin signaling pathway and restrained apoptosis via inhibiting Caspase-3 signaling pathway, whereas TRIM8 suppressed cell growth by repressing the Wnt5a/β-catenin signaling pathway and induced apoptosis through activating Caspase-3 signaling pathway. Conclusions The current study demonstrates that miR-665 facilitates LUSC cell proliferation and cell cycle transition by regulation of the Wnt5a/β-Catenin signaling pathway and represses cell apoptosis via modulation of Caspase-3 signaling pathway by directly targeting TRIM8. These findings suggest that miR-665 might be a potential new target for LUSC therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-01913-z.
Collapse
Affiliation(s)
- Tian-Jun Chen
- Respiratory Department, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Qi Zheng
- First Department of Medical Oncology, Affiliated Shaanxi Provincial Cancer Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Fei Gao
- Hua-Shan Central Hospital of Xi'an, Xi'an, 710043, People's Republic of China
| | - Tian Yang
- Respiratory Department, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Hui Ren
- Respiratory Department, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Yang Li
- Respiratory Department, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Ming-Wei Chen
- Respiratory Department, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| |
Collapse
|
6
|
Yang YCSH, Ko PJ, Pan YS, Lin HY, Whang-Peng J, Davis PJ, Wang K. Role of thyroid hormone-integrin αvβ3-signal and therapeutic strategies in colorectal cancers. J Biomed Sci 2021; 28:24. [PMID: 33827580 PMCID: PMC8028191 DOI: 10.1186/s12929-021-00719-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Thyroid hormone analogues-particularly, L-thyroxine (T4) has been shown to be relevant to the functions of a variety of cancers. Integrin αvβ3 is a plasma membrane structural protein linked to signal transduction pathways that are critical to cancer cell proliferation and metastasis. Thyroid hormones, T4 and to a less extend T3 bind cell surface integrin αvβ3, to stimulate the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway to stimulate cancer cell growth. Thyroid hormone analogues also engage in crosstalk with the epidermal growth factor receptor (EGFR)-Ras pathway. EGFR signal generation and, downstream, transduction of Ras/Raf pathway signals contribute importantly to tumor cell progression. Mutated Ras oncogenes contribute to chemoresistance in colorectal carcinoma (CRC); chemoresistance may depend in part on the activity of ERK1/2 pathway. In this review, we evaluate the contribution of thyroxine interacting with integrin αvβ3 and crosstalking with EGFR/Ras signaling pathway non-genomically in CRC proliferation. Tetraiodothyroacetic acid (tetrac), the deaminated analogue of T4, and its nano-derivative, NDAT, have anticancer functions, with effectiveness against CRC and other tumors. In Ras-mutant CRC cells, tetrac derivatives may overcome chemoresistance to other drugs via actions initiated at integrin αvβ3 and involving, downstream, the EGFR-Ras signaling pathways.
Collapse
Affiliation(s)
- Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, 11031, Taiwan
| | - Po-Jui Ko
- School of Medicine, I-Shou University, Kaohsiung, 84001, Taiwan.,Department of Pediatrics, E-DA Hospital, Kaohsiung, 82445, Taiwan
| | - Yi-Shin Pan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hung-Yun Lin
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan. .,Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan. .,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan. .,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan. .,Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, 12144, USA.
| | - Jacqueline Whang-Peng
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.,Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan
| | - Paul J Davis
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, 12144, USA.,Albany Medical College, Albany, NY, 12144, USA
| | - Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| |
Collapse
|
7
|
Zhou W, Mei J, Gu D, Xu J, Wang R, Wang H, Liu C. Wnt5a: A promising therapeutic target in ovarian cancer. Pathol Res Pract 2021; 219:153348. [DOI: 10.1016/j.prp.2021.153348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023]
|
8
|
Begum D, Merchant N, Nagaraju GP. Role of selected phytochemicals on gynecological cancers. A THERANOSTIC AND PRECISION MEDICINE APPROACH FOR FEMALE-SPECIFIC CANCERS 2021:1-30. [DOI: 10.1016/b978-0-12-822009-2.00001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Terraneo N, Jacob F, Dubrovska A, Grünberg J. Novel Therapeutic Strategies for Ovarian Cancer Stem Cells. Front Oncol 2020; 10:319. [PMID: 32257947 PMCID: PMC7090172 DOI: 10.3389/fonc.2020.00319] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is one of the most lethal gynecologic malignancies. Due to the lack of specific symptoms and screening methods, this disease is usually diagnosed only at an advanced and metastatic stage. The gold-standard treatment for OC patients consists of debulking surgery followed by taxane combined with platinum-based chemotherapy. Most patients show complete clinical remission after first-line therapy, but the majority of them ultimately relapse, developing radio- and chemoresistant tumors. It is now proposed that the cause of recurrence and reduced therapy efficacy is the presence of small populations of cancer stem cells (CSCs). These cells are usually resistant against conventional cancer therapies and for this reason, effective targeted therapies for the complete eradication of CSCs are urgently needed. In this review article, we highlight the mechanisms of CSC therapy resistance, epithelial-to-mesenchymal transition, stemness, and novel therapeutic strategies for ovarian CSCs.
Collapse
Affiliation(s)
- Nastassja Terraneo
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
| | - Francis Jacob
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Jürgen Grünberg
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
| |
Collapse
|
10
|
Asem M, Young AM, Oyama C, Claure De La Zerda A, Liu Y, Yang J, Hilliard TS, Johnson J, Harper EI, Guldner I, Zhang S, Page-Mayberry T, Kaliney WJ, Stack MS. Host Wnt5a Potentiates Microenvironmental Regulation of Ovarian Cancer Metastasis. Cancer Res 2020; 80:1156-1170. [PMID: 31932454 PMCID: PMC8245162 DOI: 10.1158/0008-5472.can-19-1601] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/09/2019] [Accepted: 01/09/2020] [Indexed: 12/19/2022]
Abstract
The noncanonical Wnt ligand Wnt5a is found in high concentrations in ascites of women with ovarian cancer. In this study, we elucidated the role of Wnt5a in ovarian cancer metastasis. Wnt5a promoted ovarian tumor cell adhesion to peritoneal mesothelial cells as well as migration and invasion, leading to colonization of peritoneal explants. Host components of the ovarian tumor microenvironment, notably peritoneal mesothelial cells and visceral adipose, secreted Wnt5a. Conditional knockout of host WNT5A significantly reduced peritoneal metastatic tumor burden. Tumors formed in WNT5A knockout mice had elevated cytotoxic T cells, increased M1 macrophages, and decreased M2 macrophages, indicating that host Wnt5a promotes an immunosuppressive microenvironment. The Src family kinase Fgr was identified as a downstream effector of Wnt5a. These results highlight a previously unreported role for host-expressed Wnt5a in ovarian cancer metastasis and suggest Fgr as a novel target for inhibition of ovarian cancer metastatic progression.Significance: This study establishes host-derived Wnt5a, expressed by peritoneal mesothelial cells and adipocytes, as a primary regulator of ovarian cancer intraperitoneal metastatic dissemination and identifies Fgr kinase as novel target for inhibition of metastasis.
Collapse
Affiliation(s)
- Marwa Asem
- Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, Indiana
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Allison M Young
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Carlysa Oyama
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | | | - Yueying Liu
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Jing Yang
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Tyvette S Hilliard
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Jeffery Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Elizabeth I Harper
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Ian Guldner
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
- Department of Biological Sciences, University of Notre Dame; South Bend, Indiana
| | - Siyuan Zhang
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
- Department of Biological Sciences, University of Notre Dame; South Bend, Indiana
| | - Toni Page-Mayberry
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - William J Kaliney
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - M Sharon Stack
- Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, Indiana.
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| |
Collapse
|
11
|
Regulation of carcinogenesis and modulation through Wnt/β-catenin signaling by curcumin in an ovarian cancer cell line. Sci Rep 2019; 9:17267. [PMID: 31754130 PMCID: PMC6872918 DOI: 10.1038/s41598-019-53509-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/31/2019] [Indexed: 12/24/2022] Open
Abstract
The secreted frizzled-related protein 5 gene (SFRP5) that antagonize the Wnt/β-catenin signaling is frequently inactivated by promoter methylation and oncogenic activation of the Wnt signaling pathway is common in many cancers. The curcumin-rich Curcuma longa has been reported to potent anti-cancer property involved in epigenetic regulation to inhibit tumor suppressor gene methylation and re-expression. In a compounds screening, we found that curcumin can inhibit Wnt/β-catenin signaling. Therefore, the aim of this study was to investigate the effects of curcumin on SFRP5 DNA methylation modification in an ovarian cancer cell line (SKOV3). SKOV3 cells were treated with DMSO, 10 μM 5-aza-2′-deoxycytidine (DAC), 5 μM DAC, 20 μM curcumin, and 20 μM curcumin combined with 5 μM DAC for 96 hours, following which RNA and proteins were extracted for further analysis. The results showed that curcumin combined with 5 μM DAC may inhibit cancer cell colony formation, migration through EMT (epithelial–mesenchymal transition) process regulation, total DNMT activity, especially in DNMT3a protein expression, and may also regulate tumor suppressor gene SFRP5 expression involved in the Wnt/β-catenin signaling pathway. The combined treatment attenuated ovarian cancer development.
Collapse
|
12
|
Wang Y, Cao J, Liu W, Zhang J, Wang Z, Zhang Y, Hou L, Chen S, Hao P, Zhang L, Zhuang M, Yu Y, Li D, Fan G. Protein tyrosine phosphatase receptor type R (PTPRR) antagonizes the Wnt signaling pathway in ovarian cancer by dephosphorylating and inactivating β-catenin. J Biol Chem 2019; 294:18306-18323. [PMID: 31653698 DOI: 10.1074/jbc.ra119.010348] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/15/2019] [Indexed: 11/06/2022] Open
Abstract
Despite a lack of mutations, accumulating evidence supports an important role for the Wnt/β-catenin pathway in ovarian tumorigenesis. However, the molecular mechanism that contributes to the aberrant activation of the Wnt signaling cascade in ovarian cancer has not been fully elucidated. Here, we found that protein tyrosine phosphatase receptor type R (PTPRR) suppressed the activation of the Wnt/β-catenin pathway in ovarian cancer. We performed an shRNA-based biochemical screen, which identified PTPRR as being responsible for tyrosine dephosphorylation of β-catenin on Tyr-142, a key site controlling the transcriptional activity of β-catenin. Of note, PTPRR was down-regulated in ovarian cancers, and ectopic PTPRR re-expression delayed ovarian cancer cell growth both in vitro and in vivo Using a proximity-based tagging system and RNA-Seq analysis, we identified a signaling nexus that includes PTPRR, α-catenin, β-catenin, E-cadherin, and AT-rich interaction domain 3C (ARID3C) in ovarian cancer. Immunohistochemistry staining of human samples further suggested that PTPRR expression is inversely correlated with disease prognosis. Collectively, our findings indicate that PTPRR functions as a tumor suppressor in ovarian cancer by dephosphorylating and inactivating β-catenin. These results suggest that PTPRR expression might have utility as a prognostic marker for predicting overall survival.
Collapse
Affiliation(s)
- Yuetong Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jian Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Weiwei Liu
- Institute of Biophysics, Key Laboratory of RNA Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jiali Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zuo Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yiqun Zhang
- Institute of Biophysics, Key Laboratory of RNA Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Linjun Hou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shengmiao Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Liye Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Min Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yang Yu
- Institute of Biophysics, Key Laboratory of RNA Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Dake Li
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China.
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
13
|
Gu ZW, He YF, Wang WJ, Tian Q, Di W. MiR-1180 from bone marrow-derived mesenchymal stem cells induces glycolysis and chemoresistance in ovarian cancer cells by upregulating the Wnt signaling pathway. J Zhejiang Univ Sci B 2019; 20:219-237. [PMID: 30829010 DOI: 10.1631/jzus.b1800190] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Bone marrow-derived mesenchymal stem cells (BM-MSCs) play an important role in cancer development and progression. However, the mechanism by which they enhance the chemoresistance of ovarian cancer is unknown. METHODS Conditioned media of BM-MSCs (BM-MSC-CM) were analyzed using a technique based on microRNA arrays. The most highly expressed microRNAs were selected for testing their effects on glycolysis and chemoresistance in SKOV3 and COC1 ovarian cancer cells. The targeted gene and related signaling pathway were investigated using in silico analysis and in vitro cancer cell models. Kaplan-Merier survival analysis was performed on a population of 59 patients enrolled to analyze the clinical significance of microRNA findings in the prognosis of ovarian cancer. RESULTS MiR-1180 was the most abundant microRNA detected in BM-MSC-CM, which simultaneously induces glycolysis and chemoresistance (against cisplatin) in ovarian cancer cells. The secreted frizzled-related protein 1 (SFRP1) gene was identified as a major target of miR-1180. The overexpression of miR-1180 led to the activation of Wnt signaling and its downstream components, namely Wnt5a, β-catenin, c-Myc, and CyclinD1, which are responsible for glycolysis-induced chemoresistance. The miR-1180 level was inversely correlated with SFRP1 mRNA expression in ovarian cancer tissue. The overexpressed miR-1180 was associated with a poor prognosis for the long-term (96-month) survival of ovarian cancer patients. CONCLUSIONS BM-MSCs enhance the chemoresistance of ovarian cancer by releasing miR-1180. The released miR-1180 activates the Wnt signaling pathway in cancer cells by targeting SFRP1. The enhanced Wnt signaling upregulates the glycolytic level (i.e. Warburg effect), which reinforces the chemoresistance property of ovarian cancer cells.
Collapse
Affiliation(s)
- Zhuo-Wei Gu
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yi-Feng He
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wen-Jing Wang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qi Tian
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wen Di
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
14
|
Guan D, Li C, Lv X, Yang Y. Pseudolaric acid B inhibits PAX2 expression through Wnt signaling and induces BAX expression, therefore promoting apoptosis in HeLa cervical cancer cells. J Gynecol Oncol 2019; 30:e77. [PMID: 31328459 PMCID: PMC6658601 DOI: 10.3802/jgo.2019.30.e77] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/11/2019] [Accepted: 03/05/2019] [Indexed: 01/05/2023] Open
Abstract
Objectives Pseudolaric acid B (PAB) has been shown to inhibit the growth of various tumor cells, but the molecular details of its function are still unknown. This study investigated the molecular mechanisms by which PAB induces apoptosis in HeLa cells. Methods The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays were performed to investigate the effect of PAB treatment in various cervical cancer cell lines. Annexin V/propidium iodide staining combined with flow cytometry and Hoechst 33258 staining were used to assess PAB-induced apoptosis. Additionally, we performed bioinformatics analyses and identified a paired box 2 (PAX2) binding site on the BAX promoter. We then validated the binding using luciferase and chromatin immunoprecipitation assays. Finally, western blotting assays were used to investigate PAB effect on the Wnt signaling and the involved signaling molecules. Results PAB promotes apoptosis and downregulates PAX2 expression in HeLa cells in a time- and concentration-dependent manner. PAX2 binds to the promoter of BAX and inhibits its expression; therefore, PAX2 inhibition is associated with increased levels of BAX, which induces apoptosis of HeLa cells via the mitochondrial pathway. Additionally, PAB inhibits classical Wnt signaling. Conclusion PAB effectively inhibits Wnt signaling and PAX2 expression, and increases BAX levels, which induce apoptosis in HeLa cells. Therefore, PAB is a promising natural molecule for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Defeng Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Chenyang Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Xiao Lv
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yongxiu Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China.,Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
15
|
Raghavan S, Mehta P, Xie Y, Lei YL, Mehta G. Ovarian cancer stem cells and macrophages reciprocally interact through the WNT pathway to promote pro-tumoral and malignant phenotypes in 3D engineered microenvironments. J Immunother Cancer 2019; 7:190. [PMID: 31324218 PMCID: PMC6642605 DOI: 10.1186/s40425-019-0666-1] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/07/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Innate immune cells such as macrophages are abundantly present within malignant ascites, where they share the microenvironment with ovarian cancer stem cells (CSC). METHODS To mimic this malignant ascites microenvironment, we created a hanging-drop hetero-spheroid model to bring CSCs and macrophages in close association. Within these hetero-spheroids, CD68+ macrophages (derived from U937 or peripheral blood monocytes) make up ~ 20% of the population, while the rest are ovarian cancer cells and ovarian cancer stem cells (derived from the high grade serous ovarian cancer cell line, OVCAR3). RESULTS Our results indicate that CSCs drive the upregulation of M2 macrophage marker CD206 within hetero-spheroids, compared to bulk ovarian cancer cells, implying an inherently more immuno-suppressive program. Moreover, an increased maintenance of elevated aldehyde dehydrogenase (ALDH) activity is noted within hetero-spheroids that include pre-polarized CD206+ M2 macrophages, implying a reciprocal interaction that drives pro-tumoral activation as well as CSC self-renewal. Consistent with enriched CSCs, we also observe increased levels of pro-tumoral IL-10 and IL-6 cytokines in the CSC/M2-macrophage hetero-spheroids. CSC/M2-macrophage hetero-spheroids are also less sensitive to the chemotherapeutic agent carboplatin and are subsequently more invasive in transwell assays. Using inhibitors of WNT secretion in both CSCs and macrophages, we found that CSC-derived WNT ligands drove CD206+ M2 macrophage activation, and that, conversely, macrophage-derived WNT ligands enriched ALDH+ cells within the CSC compartment of hetero-spheroids. Upon examination of specific WNT ligand expression within the monocyte-derived macrophage system, we observed a significant elevation in gene expression for WNT5B. In CSCs co-cultured with macrophages within hetero-spheroids, increases in several WNT ligands were observed, and this increase was significantly inhibited when WNT5B was knocked down in macrophages. CONCLUSIONS Our data implies that macrophage- initiated WNT signaling could play a significant role in the maintenance of stemness, and the resulting phenotypes of chemoresistance and invasiveness. Our results indicate paracrine WNT activation during CSC/M2 macrophages interaction constitutes a positive feedback loop that likely contributes to the more aggressive phenotype, which makes the WNT pathway a potential target to reduce the CSC and M2 macrophage compartments in the tumor microenvironment.
Collapse
Affiliation(s)
- Shreya Raghavan
- Department of Materials Science and Engineering, 2800 Plymouth Rd, Building 28, Room 3044W, Ann Arbor, MI, 48109, USA
| | - Pooja Mehta
- Department of Materials Science and Engineering, 2800 Plymouth Rd, Building 28, Room 3044W, Ann Arbor, MI, 48109, USA
| | - Yuying Xie
- Department of Computational Mathematics, Science, and Engineering, Michigan State University, East Lansing, MI, 48823, USA
| | - Yu L Lei
- Department of Periodontics and Oral Medicine and Department of Otolaryngology Head and Neck Surgery, Ann Arbor, USA
- Rogel Cancer Center, North Campus Research Complex, University of Michigan, 2800 Plymouth Rd, Building 28, Room 3044W, Ann Arbor, MI, 48109, USA
| | - Geeta Mehta
- Department of Materials Science and Engineering, 2800 Plymouth Rd, Building 28, Room 3044W, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, 2800 Plymouth Rd, Building 28, Room 3044W, Ann Arbor, MI, 48109, USA.
- Department of Macromolecular Sciences and Engineering, 2800 Plymouth Rd, Building 28, Room 3044W, Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, North Campus Research Complex, University of Michigan, 2800 Plymouth Rd, Building 28, Room 3044W, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
16
|
Bocchicchio S, Tesone M, Irusta G. Convergence of Wnt and Notch signaling controls ovarian cancer cell survival. J Cell Physiol 2019; 234:22130-22143. [PMID: 31087357 DOI: 10.1002/jcp.28775] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/10/2019] [Accepted: 04/17/2019] [Indexed: 12/20/2022]
Abstract
In the last 40 years ovarian cancer mortality rates have slightly declined and, consequently, it continues to be the fifth cause of cancer death in women. In the present study, we showed that β-catenin signaling is involved in the functions of ovarian cancer cells and interacts with the Notch system. Wnt and Notch systems showed to be prosurvival for ovarian cancer cells and their inhibition impaired cell proliferation and migration. We also demonstrated that the inhibition of β-catenin by means of two molecules, XAV939 and ICG-001, decreased the proliferation of the IGROV1 and SKOV3 ovarian cancer cell lines and that ICG-001 increased the percentage of IGROV1 cells undergoing apoptosis. The simultaneous inhibition of β-catenin and Notch signaling, by using the DAPT inhibitor, decreased ovarian cancer cell proliferation to the same extent as targeting only the Wnt/β-catenin pathway. A similar effect was observed in IGROV1 cell migration with ICG-001 and DAPT. ICG-001 increased the Notch target genes Hes-1 and Hey-1 and increased Jagged1 expression. However, no changes were observed in Dll4 or Notch 1 and 4 expressions. Our results suggest that Notch and β-catenin signaling co-operate in ovarian cancer to ensure the proliferation and migration of cells and that this could be achieved, at least partly, by the upregulation of Notch Jagged1 ligand in the absence of Wnt signaling. We showed that the Wnt pathway crosstalks with Notch in ovarian cancer cell functions, which may have implications in ovarian cancer therapeutics.
Collapse
Affiliation(s)
- Sebastián Bocchicchio
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
17
|
Chang TC, Chin YT, Nana AW, Wang SH, Liao YM, Chen YR, Shih YJ, Changou CA, Yang YCS, Wang K, Whang-Peng J, Wang LS, Stain SC, Shih A, Lin HY, Wu CH, Davis PJ. Enhancement by Nano-Diamino-Tetrac of Antiproliferative Action of Gefitinib on Colorectal Cancer Cells: Mediation by EGFR Sialylation and PI3K Activation. Discov Oncol 2018; 9:420-432. [PMID: 30187356 PMCID: PMC6223990 DOI: 10.1007/s12672-018-0341-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023] Open
Abstract
Drug resistance complicates the clinical use of gefitinib. Tetraiodothyroacetic acid (tetrac) and nano-diamino-tetrac (NDAT) have been shown in vitro and in xenografts to have antiproliferative/angiogenic properties and to potentiate antiproliferative activity of other anticancer agents. In the current study, we investigated the effects of NDAT on the anticancer activities of gefitinib in human colorectal cancer cells. β-Galactoside α-2,6-sialyltransferase 1 (ST6Gal1) catalyzes EGFR sialylation that is associated with gefitinib resistance in colorectal cancers, and this was also investigated. Gefitinib inhibited cell proliferation of HT-29 cells (K-ras wild-type), and NDAT significantly enhanced the antiproliferative action of gefitinib. Gefitinib inhibited cell proliferation of HCT116 cells (K-ras mutant) only in high concentration, and this was further enhanced by NDAT. NDAT enhancedd gefitinib-induced antiproliferation in gefitinib-resistant colorectal cancer cells by inhibiting ST6Gal1 activity and PI3K activation. Furthermore, NDAT enhanced gefitinib-induced anticancer activity additively in colorectal cancer HCT116 cell xenograft-bearing nude mice. Results suggest that NDAT may have an application with gefitinib as combination colorectal cancer therapy.
Collapse
Affiliation(s)
- Tung-Cheng Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Division of Colorectal Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.,Division of Colorectal Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Tang Chin
- Taipei Cancer Center, Taipei Medical University, Taipei, 11031, Taiwan.,The PhD program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - André Wendindondé Nana
- The PhD program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Shwu-Huey Wang
- Core Facility Center, Office of Research and Development, Taipei Medical University, Taipei, 11031, Taiwan.,Department of Biochemistry and Molecular Cell Biology, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Min Liao
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Yi-Ru Chen
- Taipei Cancer Center, Taipei Medical University, Taipei, 11031, Taiwan.,The PhD program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ya-Jung Shih
- Taipei Cancer Center, Taipei Medical University, Taipei, 11031, Taiwan.,The PhD program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chun A Changou
- The PhD program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Core Facility Center, Office of Research and Development, Taipei Medical University, Taipei, 11031, Taiwan.,Integrated Laboratory, Center of Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Chen Sh Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Jacqueline Whang-Peng
- Taipei Cancer Center; Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Liang-Shun Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Department of Surgery, Shuang Ho Hospital, Taipei Medical University, No. 291, Zhongzheng Rd., Zhonghe, New Taipei City, 23561, Taiwan
| | - Steven C Stain
- Department of Surgery, Albany Medical College, Albany, NY, 12208, USA
| | - Ai Shih
- National Laboratory Animal Center, Taipei, 11599, Taiwan
| | - Hung-Yun Lin
- Taipei Cancer Center, Taipei Medical University, Taipei, 11031, Taiwan. .,The PhD program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan. .,Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, 12144, USA. .,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan. .,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Chih-Hsiung Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan. .,Department of Surgery, Shuang Ho Hospital, Taipei Medical University, No. 291, Zhongzheng Rd., Zhonghe, New Taipei City, 23561, Taiwan.
| | - Paul J Davis
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, 12144, USA. .,NanoPharmaceuticals LLC, Rensselaer, NY, 12144, USA. .,Department of Medicine, Albany Medical College, Albany, NY, 12208, USA.
| |
Collapse
|
18
|
Cao P, Zhao S, Sun Z, Jiang N, Shang Y, Wang Y, Gu J, Li S. BRMS1L suppresses ovarian cancer metastasis via inhibition of the β-catenin-wnt pathway. Exp Cell Res 2018; 371:214-221. [PMID: 30118697 DOI: 10.1016/j.yexcr.2018.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 01/19/2023]
Abstract
A low level of breast cancer metastasis suppressor 1-like (BRMS1L) has been implicated in tumour metastasis involving breast cancer and other cancers. It remains unclear whether BRMS1L is involved in epithelial ovarian cancer (EOC) metastasis and what the molecular mechanism of BRMS1L is in suppressing EOC metastasis. In this study, we examined the mRNA expression and protein level of BRMS1L by screening EOC patients. Our results show that BRMS1L expression is downregulated in EOC patients compared to that in normal people and negatively correlated to pathological stages of EOC. We further explored examining epithelial to mesenchymal transition (EMT) as the molecular mechanism of BRMS1L in cancer cell metastasis. The overexpression of BRMS1L inhibits EOC cell migration and invasion, and this inhibition is correlated to the inactivation of EMT and Wnt/β-catenin signalling in vitro. Knockdown of BRMS1L by shRNA promotes EOC metastasis, enhances EMT process and activates Wnt/β-catenin signalling. These results suggest that BRMS1L plays a critical role in the suppression of ovarian cancer metastasis, and BRMS1L can be considered as a prognostic biomarker and potential therapeutic target for EOC patients.
Collapse
Affiliation(s)
- Penglong Cao
- Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Shuai Zhao
- Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Zhigang Sun
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Nan Jiang
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yuhong Shang
- Department of Gynecology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yingxin Wang
- Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Juebin Gu
- Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Shijun Li
- Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
19
|
Can Stemness and Chemoresistance Be Therapeutically Targeted via Signaling Pathways in Ovarian Cancer? Cancers (Basel) 2018; 10:cancers10080241. [PMID: 30042330 PMCID: PMC6116003 DOI: 10.3390/cancers10080241] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/12/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy. Poor overall survival, particularly for patients with high grade serous (HGS) ovarian cancer, is often attributed to late stage at diagnosis and relapse following chemotherapy. HGS ovarian cancer is a heterogenous disease in that few genes are consistently mutated between patients. Additionally, HGS ovarian cancer is characterized by high genomic instability. For these reasons, personalized approaches may be necessary for effective treatment and cure. Understanding the molecular mechanisms that contribute to tumor metastasis and chemoresistance are essential to improve survival rates. One favored model for tumor metastasis and chemoresistance is the cancer stem cell (CSC) model. CSCs are cells with enhanced self-renewal properties that are enriched following chemotherapy. Elimination of this cell population is thought to be a mechanism to increase therapeutic response. Therefore, accurate identification of stem cell populations that are most clinically relevant is necessary. While many CSC identifiers (ALDH, OCT4, CD133, and side population) have been established, it is still not clear which population(s) will be most beneficial to target in patients. Therefore, there is a critical need to characterize CSCs with reliable markers and find their weaknesses that will make the CSCs amenable to therapy. Many signaling pathways are implicated for their roles in CSC initiation and maintenance. Therapeutically targeting pathways needed for CSC initiation or maintenance may be an effective way of treating HGS ovarian cancer patients. In conclusion, the prognosis for HGS ovarian cancer may be improved by combining CSC phenotyping with targeted therapies for pathways involved in CSC maintenance.
Collapse
|
20
|
Nerve growth factor modulates the tumor cells migration in ovarian cancer through the WNT/β-catenin pathway. Oncotarget 2018; 7:81026-81048. [PMID: 27835587 PMCID: PMC5348374 DOI: 10.18632/oncotarget.13186] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/21/2016] [Indexed: 12/21/2022] Open
Abstract
Nerve growth factor (NGF)/nerve growth factor receptors (NGFRs) axis and canonical WNT/β-catenin pathway have shown to play crucial roles in tumor initiation, progression and prognosis. But little did we know the relationship between them in modulation of tumor progress. In this report, we found that NGF/NGFRs and β-catenin were coexpression in ovarian cancer cell lines, and NGF can decrease the expression level of β-catenin and affect its activities, which may be related to the NGF-induced down-regulation of B-cell CLL/lymphoma 9-like (BCL9L, BCL9-2). Furthermore, NGF can also increase or decrease the downstream target gene expression levels of WNT/β-catenin depending on the cell types. Especially, we created a novel in vitro cell growth model based on a microfluidic device to intuitively observe the effects of NGF/NGFRs on the motility behaviors of ovarian cancer cells. The results showed that the migration area and maximum distance into three dimensional (3D) matrigel were decreased in CAOV3 and OVCAR3 cells, but increased in SKOV3 cells following the stimulation with NGF. In addition, we found that the cell colony area was down-regulated in CAOV3 cells, however, it was augmented in OVCAR3 cells after treatment with NGF. The inhibitors of NGF/NGFRs, such as Ro 08-2750, K252a and LM11A-31,can all block NGF-stimulated changes of gene expression or migratory behavior on ovarian cancer cells. The different results among ovarian cancer cells illustrated the heterogeneity and complexity of ovarian cancer. Collectively, our results suggested for the first time that NGF is functionally linked to β-catenin in the migration of human ovarian cancer cells, which may be a novel therapeutic perspective to prevent the spread of ovarian carcinomas by studying the interaction between NGF/NGFRs and canonical WNT/β-catenin signaling.
Collapse
|
21
|
Kwon M, Kim JH, Rybak Y, Luna A, Choi CH, Chung JY, Hewitt SM, Adem A, Tubridy E, Lin J, Libutti SK. Reduced expression of FILIP1L, a novel WNT pathway inhibitor, is associated with poor survival, progression and chemoresistance in ovarian cancer. Oncotarget 2018; 7:77052-77070. [PMID: 27776341 PMCID: PMC5340232 DOI: 10.18632/oncotarget.12784] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/17/2016] [Indexed: 12/15/2022] Open
Abstract
Filamin A interacting protein 1-like (FILIP1L) is an inhibitor of the canonical WNT pathway. WNT/β-catenin signaling and its downstream pathway, epithelial-to-mesenchymal transition (EMT), play a key role in ovarian cancer metastasis and chemoresistance. To study the clinical implications of FILIP1L in regulating the WNT/β-catenin pathway, the expression of FILIP1L, β-catenin, SNAIL and SLUG was analyzed by immunohistochemistry on tissue microarrays of 369 ovarian samples ranging from normal to metastatic. In addition, the results were validated in mouse model and in vitro cell culture. In the present study, we demonstrated that FILIP1L expression was inversely correlated with poor prognosis, stage and chemoresistance in ovarian cancer. Notably, low FILIP1L expression was independent negative prognostic factor with respect to overall and disease-free survival. FILIP1L inhibited peritoneal metastases in orthotopic mouse model. FILIP1L knockdown induced chemoresistance in ovarian cancer cells and this phenotype was rescued by simultaneous knockdown of FILIP1L and SLUG, an EMT activator. We also demonstrated that FILIP1L regulates β-catenin degradation. FILIP1L co-localizes with phospho-β-catenin and increases phospho-β-catenin at the centrosomes, destined for proteosomal degradation. Finally, we showed that FILIP1L regulates EMT. Overall, these findings suggest that FILIP1L promotes β-catenin degradation and suppresses EMT, thereby inhibiting metastases and chemoresistance. Our study provides the first clinical relevance of FILIP1L in human cancer, and suggests that FILIP1L may be a novel prognostic marker for chemotherapy in ovarian cancer patients. Further, the modulation of FILIP1L expression may have the potential to be a target for cancer therapy.
Collapse
Affiliation(s)
- Mijung Kwon
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 135-720, Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 135-720, Korea
| | - Yevangelina Rybak
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Alex Luna
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea.,Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joon-Yong Chung
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen M Hewitt
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Asha Adem
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Elizabeth Tubridy
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Juan Lin
- Division of Biostatistics, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Steven K Libutti
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
22
|
Niiro E, Morioka S, Iwai K, Yamada Y, Ogawa K, Kawahara N, Kobayashi H. Potential signaling pathways as therapeutic targets for overcoming chemoresistance in mucinous ovarian cancer. Biomed Rep 2018; 8:215-223. [PMID: 29564122 DOI: 10.3892/br.2018.1045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 01/10/2018] [Indexed: 12/14/2022] Open
Abstract
Cases of mucinous ovarian cancer are predominantly resistant to chemotherapies. The present review summarizes current knowledge of the therapeutic potential of targeting the Wingless (WNT) pathway, with particular emphasis on preclinical and clinical studies, for improving the chemoresistance and treatment of mucinous ovarian cancer. A review was conducted of English language literature published between January 2000 and October 2017 that concerned potential signaling pathways associated with the chemoresistance of mucinous ovarian cancer. The literature indicated that aberrant activation of growth factor and WNT signaling pathways is specifically observed in mucinous ovarian cancer. An evolutionarily conserved signaling cascade system including epidermal growth factor/RAS/RAF/mitogen-activated protein kinase kinase/extracellular signal-regulated protein kinase, phosphoinositide 3-kinase/Akt and WNT signaling regulates a variety of cellular functions; their crosstalk mutually enhances signaling activity and induces chemoresistance. Novel antagonists, modulators and inhibitors have been developed for targeting the components of the WNT signaling pathway, namely Frizzled, low-density lipoprotein receptor-related protein 5/6, Dishevelled, casein kinase 1, AXIN, glycogen synthase kinase 3β and β-catenin. Targeted inhibition of WNT signaling represents a rational and promising novel approach to overcome chemoresistance, and several WNT inhibitors are being evaluated in preclinical studies. In conclusion, the WNT receptors and their downstream components may serve as novel therapeutic targets for overcoming chemoresistance in mucinous ovarian cancer.
Collapse
Affiliation(s)
- Emiko Niiro
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Sachiko Morioka
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Kana Iwai
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Yuki Yamada
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Kenji Ogawa
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Naoki Kawahara
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
23
|
Pfankuchen DB, Baltes F, Batool T, Li JP, Schlesinger M, Bendas G. Heparin antagonizes cisplatin resistance of A2780 ovarian cancer cells by affecting the Wnt signaling pathway. Oncotarget 2017; 8:67553-67566. [PMID: 28978053 PMCID: PMC5620193 DOI: 10.18632/oncotarget.18738] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/22/2017] [Indexed: 01/02/2023] Open
Abstract
Low molecular weight heparin (LMWH), the guideline based drug for prophylaxis and treatment of cancer-associated thrombosis, was recently shown to sensitize cisplatin resistant A2780cis human ovarian cancer cells for cisplatin cytotoxicity upon 24 h pretreatment with 50 μg × mL−1 of the LMWH tinzaparin in vitro, equivalent to a therapeutic dosage. Thereby, LMWH induced sensitization by transcriptional reprogramming of A2780cis cells via not yet elucidated mechanisms that depend on cellular proteoglycans. Here we aim to illuminate the underlying molecular mechanisms of LMWH in sensitizing A2780cis cells for cisplatin. Using TCF/LEF luciferase promotor assay (Top/Flash) we show that resistant A2780cis cells possess a threefold higher Wnt signaling activity compared to A2780 cells. Furthermore, Wnt pathway blockade by FH535 leads to higher cisplatin sensitivity of A2780cis cells. Glypican-3 (GPC3) is upregulated in A2780cis cells in response to LMWH treatment, probably as counter-regulation to sustain the high Wnt activity against LMWH. Hence, LMWH reduces the cisplatin-induced rise in Wnt activity and TCF-4 expression in A2780cis cells, but keeps sensitive A2780 cells unaffected. Consequently, Wnt signaling pathway appears as primary target of LMWH in sensitizing A2780cis cells for cisplatin toxicity. Considering the outstanding role of LMWH in clinical oncology, this finding appears as promising therapeutic option to hamper chemoresistance.
Collapse
Affiliation(s)
| | - Fabian Baltes
- Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Tahira Batool
- Department of Medical Biochemistry and Microbiology, SciLifeLab, University of Uppsala, Uppsala, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, SciLifeLab, University of Uppsala, Uppsala, Sweden
| | - Martin Schlesinger
- Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Gerd Bendas
- Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| |
Collapse
|
24
|
Xu Y, Ma YH, Pang YX, Zhao Z, Lu JJ, Mao HL, Liu PS. Ectopic repression of receptor tyrosine kinase-like orphan receptor 2 inhibits malignant transformation of ovarian cancer cells by reversing epithelial-mesenchymal transition. Tumour Biol 2017; 39:1010428317701627. [PMID: 28475014 DOI: 10.1177/1010428317701627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Receptor tyrosine kinase-like orphan receptor 2 is an enzyme-linked receptor which specifically modulates WNT5A signaling and plays an important role in tumorigenesis, invasion, and metastasis; however, the precise role of receptor tyrosine kinase-like orphan receptor 2 in cancer is controversial. The purpose of this study was to investigate the expression and role of receptor tyrosine kinase-like orphan receptor 2 in ovarian carcinoma and clarify the biological functions and interactions of receptor tyrosine kinase-like orphan receptor 2 with non-canonical Wnt pathways in ovarian cancer. The result of the human ovary tissue microarray revealed that the receptor tyrosine kinase-like orphan receptor 2-positive rate increased in malignant epithelial ovarian cancers and was extremely higher in the metastatic tumor tissues, which was also higher than that in the malignant ovarian tumor tissues. In addition, high expression of receptor tyrosine kinase-like orphan receptor 2 was closely related with ovarian cancer grading. The expression of receptor tyrosine kinase-like orphan receptor 2 protein was higher in SKOV3 and A2780 cells than OVCAR3 and 3AO cells. Knockdown of receptor tyrosine kinase-like orphan receptor 2 inhibited ovarian cancer cell proliferation, migration, invasion, and induced morphologic as well as digestive state alterations in stably transfected SKOV3 cells. Detailed study further revealed that silencing of receptor tyrosine kinase-like orphan receptor 2 reversed the epithelial-mesenchymal transition and inhibited non-canonical Wnt signaling. Our findings suggest that receptor tyrosine kinase-like orphan receptor 2 may be an important regulator of epithelial-mesenchymal transition, primarily regulated the non-canonical Wnt signaling pathway in ovarian cancer cells, and may display a promising therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Ying Xu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yan-Hui Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Ying-Xin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhe Zhao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing-Jing Lu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Hong-Luan Mao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Pei-Shu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
25
|
Liu R, Cheng J, Chen Y, Wang W, Chen J, Mao G. Potential role and prognostic importance of dishevelled-2 in epithelial ovarian cancer. Int J Gynaecol Obstet 2017; 138:304-310. [PMID: 28513833 DOI: 10.1002/ijgo.12218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 03/07/2017] [Accepted: 05/15/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To investigate the role and prognostic importance of Dvl2 in human epithelial ovarian cancer (EOC). METHODS A multimethod study was undertaken including patients with pathologically confirmed non-metastatic EOC who underwent surgery for maximum tumor resection at a center in China. Dvl2 expression was assessed by western blot using fresh EOC tissues and normal ovarian tissues obtained between June 2014 and January 2015. Additionally, retrospective data were obtained for patients treated between April 2004 and September 2009. Their tumor specimens were used in immunohistochemistry analysis. Kaplan-Meier survival plots were constructed to estimate the overall survival by Dvl2 expression, and a Cox proportional hazards model was used to analyze prognostic factors. Alterations in Dvl2 expression during the cell cycle were assessed by a starvation and refeeding assay. RESULTS Dvl2 expression was higher in EOC samples than in normal tissues on western blot. Overall, 124 patients were included in immunohistochemistry analysis, and Dvl2 expression level was significantly associated with the tumor grade and Ki-67 expression. Overexpression of Dvl2 was correlated with poor prognosis. The pattern of Dvl2 expression throughout the cell cycle matched that of the cell proliferation marker cyclin D1. CONCLUSION Dvl2 could play a part in EOC progression and might be an independent prognostic factor. Additionally, it might be a prospective therapeutic target in the treatment of EOC.
Collapse
Affiliation(s)
- Rong Liu
- Department of Gynecologic Oncology, Nantong University Cancer Hospital, Nantong University, Nantong, China
| | - Jialin Cheng
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Yannan Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Wei Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Jie Chen
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.,Department of Oncology, Jiangyin People's Hospital, Wuxi, China
| | - Guoxin Mao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
26
|
Fan G, Zhang S, Gao Y, Greer PA, Tonks NK. HGF-independent regulation of MET and GAB1 by nonreceptor tyrosine kinase FER potentiates metastasis in ovarian cancer. Genes Dev 2017; 30:1542-57. [PMID: 27401557 PMCID: PMC4949327 DOI: 10.1101/gad.284166.116] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 06/07/2016] [Indexed: 12/29/2022]
Abstract
In this study, Fan et al. report a novel ligand- and autophosphorylation-independent activation of MET through the nonreceptor tyrosine kinase FER. The findings show that levels of FER were elevated in ovarian cancer cell lines and that loss of FER impaired the metastasis of ovarian cancer cells in vivo, providing new insights into signaling events that underlie metastasis in ovarian cancer cells. Ovarian cancer cells disseminate readily within the peritoneal cavity, which promotes metastasis, and are often resistant to chemotherapy. Ovarian cancer patients tend to present with advanced disease, which also limits treatment options; consequently, new therapies are required. The oncoprotein tyrosine kinase MET, which is the receptor for hepatocyte growth factor (HGF), has been implicated in ovarian tumorigenesis and has been the subject of extensive drug development efforts. Here, we report a novel ligand- and autophosphorylation-independent activation of MET through the nonreceptor tyrosine kinase feline sarcoma-related (FER). We demonstrated that the levels of FER were elevated in ovarian cancer cell lines relative to those in immortalized normal surface epithelial cells and that suppression of FER attenuated the motility and invasive properties of these cancer cells. Furthermore, loss of FER impaired the metastasis of ovarian cancer cells in vivo. Mechanistically, we demonstrated that FER phosphorylated a signaling site in MET: Tyr1349. This enhanced activation of RAC1/PAK1 and promoted a kinase-independent scaffolding function that led to recruitment and phosphorylation of GAB1 and the specific activation of the SHP2–ERK signaling pathway. Overall, this analysis provides new insights into signaling events that underlie metastasis in ovarian cancer cells, consistent with a prometastatic role of FER and highlighting its potential as a novel therapeutic target for metastatic ovarian cancer.
Collapse
Affiliation(s)
- Gaofeng Fan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Siwei Zhang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Yan Gao
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L3N6, Canada
| | - Peter A Greer
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L3N6, Canada
| | - Nicholas K Tonks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
27
|
Wnt5a Signaling in Normal and Cancer Stem Cells. Stem Cells Int 2017; 2017:5295286. [PMID: 28491097 PMCID: PMC5405594 DOI: 10.1155/2017/5295286] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/07/2017] [Indexed: 01/31/2023] Open
Abstract
Wnt5a is involved in activating several noncanonical Wnt signaling pathways, which can inhibit or activate canonical Wnt/β-catenin signaling pathway in a receptor context-dependent manner. Wnt5a signaling is critical for regulating normal developmental processes, including stem cell self-renewal, proliferation, differentiation, migration, adhesion, and polarity. Moreover, the aberrant activation or inhibition of Wnt5a signaling is emerging as an important event in cancer progression, exerting both oncogenic and tumor suppressive effects. Recent studies show the involvement of Wnt5a signaling in regulating normal and cancer stem cell self-renewal, cancer cell proliferation, migration, and invasion. In this article, we review recent findings regarding the molecular mechanisms and roles of Wnt5a signaling in stem cells in embryogenesis and in the normal or neoplastic breast or ovary, highlighting that Wnt5a may have different effects on target cells depending on the surface receptors expressed by the target cell.
Collapse
|
28
|
Lupia M, Cavallaro U. Ovarian cancer stem cells: still an elusive entity? Mol Cancer 2017; 16:64. [PMID: 28320418 PMCID: PMC5360065 DOI: 10.1186/s12943-017-0638-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/13/2017] [Indexed: 12/16/2022] Open
Abstract
The cancer stem cell (CSC) model proposes that tumor development and progression are fueled and sustained by undifferentiated cancer cells, endowed with self-renewal and tumor-initiating capacity. Ovarian carcinoma, based on its biological features and clinical evolution, appears as a prototypical example of CSC-driven disease. Indeed, ovarian cancer stem cells (OCSC) would account not only for the primary tumor growth, the peritoneal spread and the relapse, but also for the development of chemoresistance, thus having profound implication for the treatment of this deadly disease. In the last decade, an increasing body of experimental evidence has supported the existence of OCSC and their pathogenic role in the disease. Nevertheless, the identification of OCSC and the definition of their phenotypical and functional traits have proven quite challenging, mainly because of the heterogeneity of the disease and of the difficulties in establishing reliable biological models. A deeper understanding of OCSC pathobiology will shed light on the mechanisms that underlie the clinical behaviour of OC. In addition, it will favour the design of innovative treatment regimens that, on one hand, would counteract the resistance to conventional chemotherapy, and, on the other, would aim at the eradication of OC through the elimination of its CSC component.
Collapse
Affiliation(s)
- Michela Lupia
- Unit of Gynecological Oncology Research, European Institute of Oncology, Via G. Ripamonti 435, I-20141, Milan, Italy
| | - Ugo Cavallaro
- Unit of Gynecological Oncology Research, European Institute of Oncology, Via G. Ripamonti 435, I-20141, Milan, Italy.
| |
Collapse
|
29
|
Baarsma HA, Skronska-Wasek W, Mutze K, Ciolek F, Wagner DE, John-Schuster G, Heinzelmann K, Günther A, Bracke KR, Dagouassat M, Boczkowski J, Brusselle GG, Smits R, Eickelberg O, Yildirim AÖ, Königshoff M. Noncanonical WNT-5A signaling impairs endogenous lung repair in COPD. J Exp Med 2016; 214:143-163. [PMID: 27979969 PMCID: PMC5206496 DOI: 10.1084/jem.20160675] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/16/2016] [Accepted: 11/04/2016] [Indexed: 01/17/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of death worldwide. One main pathological feature of COPD is the loss of functional alveolar tissue without adequate repair (emphysema), yet the underlying mechanisms are poorly defined. Reduced WNT-β-catenin signaling is linked to impaired lung repair in COPD; however, the factors responsible for attenuating this pathway remain to be elucidated. Here, we identify a canonical to noncanonical WNT signaling shift contributing to COPD pathogenesis. We demonstrate enhanced expression of noncanonical WNT-5A in two experimental models of COPD and increased posttranslationally modified WNT-5A in human COPD tissue specimens. WNT-5A was increased in primary lung fibroblasts from COPD patients and induced by COPD-related stimuli, such as TGF-β, cigarette smoke (CS), and cellular senescence. Functionally, mature WNT-5A attenuated canonical WNT-driven alveolar epithelial cell wound healing and transdifferentiation in vitro. Lung-specific WNT-5A overexpression exacerbated airspace enlargement in elastase-induced emphysema in vivo. Accordingly, inhibition of WNT-5A in vivo attenuated lung tissue destruction, improved lung function, and restored expression of β-catenin-driven target genes and alveolar epithelial cell markers in the elastase, as well as in CS-induced models of COPD. We thus identify a novel essential mechanism involved in impaired mesenchymal-epithelial cross talk in COPD pathogenesis, which is amenable to therapy.
Collapse
Affiliation(s)
- Hoeke A Baarsma
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | - Wioletta Skronska-Wasek
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | - Kathrin Mutze
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | - Florian Ciolek
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | - Darcy E Wagner
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | - Gerrit John-Schuster
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | - Katharina Heinzelmann
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | | | - Ken R Bracke
- Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | | | | | - Guy G Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Ron Smits
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center Rotterdam, 3000 Rotterdam, Netherlands
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | - Ali Ö Yildirim
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, 81377 Munich, Germany
| |
Collapse
|
30
|
Matte I, Legault CM, Garde-Granger P, Laplante C, Bessette P, Rancourt C, Piché A. Mesothelial cells interact with tumor cells for the formation of ovarian cancer multicellular spheroids in peritoneal effusions. Clin Exp Metastasis 2016; 33:839-852. [PMID: 27612856 DOI: 10.1007/s10585-016-9821-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/31/2016] [Indexed: 12/13/2022]
Abstract
Epithelial ovarian cancer (EOC) dissemination is primarily mediated by the shedding of tumor cells from the primary site into ascites where they form multicellular spheroids that rapidly lead to peritoneal carcinomatosis. While the clinical importance and fundamental role of multicellular spheroids in EOC is increasingly appreciated, the mechanisms that regulate their formation and dictate their cellular composition remain poorly characterized. To investigate these important questions, we characterized spheroids isolated from ascites of women with EOC. We found that in these spheroids, a core of mesothelial cells was encased in a shell of tumor cells. Analysis further revealed that EOC spheroids are dynamic structures of proliferating, non-proliferating and hypoxic regions. To recapitulate these in vivo findings, we developed a three-dimensional co-culture model of primary EOC and mesothelial cells. Our analysis indicated that, compared to the OVCAR3 cell line, primary EOC cells isolated from ascites as well as mesothelial cells formed compact spheroids. Analysis of heterotypic spheroid microarchitecture revealed a structure that grossly resembles the structure of spheroids isolated from ascites. Cells that formed compact spheroids had elevated expression of β1 integrin and low expression of E-cadherin. Addition of β1 integrin blocking antibody or siRNA-mediated downregulation of β1 integrin resulted in reduced tightness of the spheroids. Interestingly, the loss of MUC16 and E-cadherin expression resulted in the formation of more compact spheroids. Therefore, our findings support the heterotypic nature of spheroids from malignant EOC ascites. In addition, our data describe an unusual link between E-cadherin expression and less compact spheroids. Our data also emphasize the role of MUC16 and β1 integrin in EOC spheroid formation.
Collapse
Affiliation(s)
- Isabelle Matte
- Département de Microbiologie et Infectiologie, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Clara Major Legault
- Département de Microbiologie et Infectiologie, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Perrine Garde-Granger
- Département de Pathologie, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Claude Laplante
- Département de Pathologie, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Paul Bessette
- Département de Chirurgie, Service d'Obstétrique-Gynécologie, Faculté de Médecine, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Claudine Rancourt
- Département de Microbiologie et Infectiologie, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Alain Piché
- Département de Microbiologie et Infectiologie, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada.
| |
Collapse
|
31
|
Wnt5a Signaling in Cancer. Cancers (Basel) 2016; 8:cancers8090079. [PMID: 27571105 PMCID: PMC5040981 DOI: 10.3390/cancers8090079] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/09/2016] [Accepted: 08/22/2016] [Indexed: 01/07/2023] Open
Abstract
Wnt5a is involved in activating several non-canonical WNT signaling pathways, through binding to different members of the Frizzled- and Ror-family receptors. Wnt5a signaling is critical for regulating normal developmental processes, including proliferation, differentiation, migration, adhesion and polarity. However, the aberrant activation or inhibition of Wnt5a signaling is emerging as an important event in cancer progression, exerting both oncogenic and tumor suppressive effects. Recent studies show the involvement of Wnt5a in regulating cancer cell invasion, metastasis, metabolism and inflammation. In this article, we review findings regarding the molecular mechanisms and roles of Wnt5a signaling in various cancer types, and highlight Wnt5a in ovarian cancer.
Collapse
|
32
|
Nagaraj AB, Joseph P, Kovalenko O, Singh S, Armstrong A, Redline R, Resnick K, Zanotti K, Waggoner S, DiFeo A. Critical role of Wnt/β-catenin signaling in driving epithelial ovarian cancer platinum resistance. Oncotarget 2016; 6:23720-34. [PMID: 26125441 PMCID: PMC4695147 DOI: 10.18632/oncotarget.4690] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/01/2015] [Indexed: 01/06/2023] Open
Abstract
Resistance to platinum-based chemotherapy is the major barrier to treating epithelial ovarian cancer. To improve patient outcomes, it is critical to identify the underlying mechanisms that promote platinum resistance. Emerging evidence supports the concept that platinum-based therapies are able to eliminate the bulk of differentiated cancer cells, but are unable to eliminate cancer initiating cells (CIC). To date, the relevant pathways that regulate ovarian CICs remain elusive. Several correlative studies have shown that Wnt/β-catenin pathway activation is associated with poor outcomes in patients with high-grade serous ovarian cancer (HGSOC). However, the functional relevance of these findings remain to be delineated. We have uncovered that Wnt/β-catenin pathway activation is a critical driver of HGSOC chemotherapy resistance, and targeted inhibition of this pathway, which eliminates CICs, represents a novel and effective treatment for chemoresistant HGSOC. Here we show that Wnt/β-catenin signaling is activated in ovarian CICs, and targeted inhibition of β-catenin potently sensitized cells to cisplatin and decreased CIC tumor sphere formation. Furthermore, the Wnt/β-catenin specific inhibitor iCG-001 potently sensitized cells to cisplatin and decreased stem-cell frequency in platinum resistant cells. Taken together, our data is the first report providing evidence that the Wnt/β-catenin signaling pathway maintains stem-like properties and drug resistance of primary HGSOC PDX derived platinum resistant models, and therapeutic targeting of this pathway with iCG-001/PRI-724, which has been shown to be well tolerated in Phase I trials, may be an effective treatment option.
Collapse
Affiliation(s)
- Anil Belur Nagaraj
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Peronne Joseph
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Olga Kovalenko
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Sareena Singh
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Amy Armstrong
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Raymond Redline
- Department of Pathology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Kimberly Resnick
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Kristine Zanotti
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Steven Waggoner
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Analisa DiFeo
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
33
|
Koussounadis A, Langdon SP, Um I, Kay C, Francis KE, Harrison DJ, Smith VA. Dynamic modulation of phosphoprotein expression in ovarian cancer xenograft models. BMC Cancer 2016; 16:205. [PMID: 26964739 PMCID: PMC4787009 DOI: 10.1186/s12885-016-2212-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/24/2016] [Indexed: 11/10/2022] Open
Abstract
Background The dynamic changes that occur in protein expression after treatment of a cancer in vivo are poorly described. In this study we measure the effect of chemotherapy over time on the expression of a panel of proteins in ovarian cancer xenograft models. The objective was to identify phosphoprotein and other protein changes indicative of pathway activation that might link with drug response. Methods Two xenograft models, platinum-responsive OV1002 and platinum-unresponsive HOX424, were used. Treatments were carboplatin and carboplatin-paclitaxel. Expression of 49 proteins over 14 days post treatment was measured by quantitative immunofluorescence and analysed by AQUA. Results Carboplatin treatment in the platinum-sensitive OV1002 model triggered up-regulation of cell cycle, mTOR and DDR pathways, while at late time points WNT, invasion, EMT and MAPK pathways were modulated. Estrogen receptor-alpha (ESR1) and ERBB pathways were down-regulated early, within 24 h from treatment administration. Combined carboplatin-paclitaxel treatment triggered a more extensive response in the OV1002 model modulating expression of 23 of 49 proteins. Therefore the cell cycle and DDR pathways showed similar or more pronounced changes than with carboplatin alone. In addition to expression of pS6 and pERK increasing, components of the AKT pathway were modulated with pAKT increasing while its regulator PTEN was down-regulated early. WNT signaling, EMT and invasion markers were modulated at later time points. Additional pathways were also observed with the NFκB and JAK/STAT pathways being up-regulated. ESR1 was down-regulated as was HER4, while further protein members of the ERBB pathway were upregulated late. By contrast, in the carboplatin-unresponsive HOX 424 xenograft, carboplatin only modulated expression of MLH1 while carboplatin-paclitaxel treatment modulated ESR1 and pMET. Conclusions Thirteen proteins were modulated by carboplatin and a more robust set of changes by carboplatin-paclitaxel. Early changes included DDR and cell cycle regulatory proteins associating with tumor volume changes, as expected. Changes in ESR1 and ERBB signaling were also observed. Late changes included components of MAPK signaling, EMT and invasion markers and coincided in time with reversal in tumor volume reduction. These results suggest potential therapeutic roles for inhibitors of such pathways that may prolong chemotherapeutic effects. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2212-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Antonis Koussounadis
- School of Biology, Sir Harold Mitchell Building, University of St Andrews, St Andrews, Fife, KY16 9TH, UK
| | - Simon P Langdon
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Inhwa Um
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.,School of Medicine, University of St Andrews, St Andrews, UK
| | - Charlene Kay
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Kyle E Francis
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | | | - V Anne Smith
- School of Biology, Sir Harold Mitchell Building, University of St Andrews, St Andrews, Fife, KY16 9TH, UK.
| |
Collapse
|
34
|
Boone JD, Arend RC, Johnston BE, Cooper SJ, Gilchrist SA, Oelschlager DK, Grizzle WE, McGwin G, Gangrade A, Straughn JM, Buchsbaum DJ. Targeting the Wnt/β-catenin pathway in primary ovarian cancer with the porcupine inhibitor WNT974. J Transl Med 2016; 96:249-59. [PMID: 26658453 DOI: 10.1038/labinvest.2015.150] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/30/2015] [Accepted: 11/09/2015] [Indexed: 01/18/2023] Open
Abstract
Preclinical studies in ovarian cancer have demonstrated upregulation of the Wnt/β-catenin pathway promoting tumor proliferation and chemoresistance. Our objective was to evaluate the effect of the Wnt/β-catenin pathway inhibitor, WNT974, in primary ovarian cancer ascites cells. Ascites cells from patients with papillary serous ovarian cancer were isolated and treated with 1 μM WNT974±100 μM carboplatin. Viability was evaluated with the ATPlite assay. The IC50 was calculated using a dose-response analysis. Immunohistochemistry (IHC) was performed on ascites cells and tumor. Expression of R-spondin 2 (RSPO2), RSPO3, PORCN, WLS, AXIN2, and three previously characterized RSPO fusion transcripts were assessed using Taqman assays. Sixty ascites samples were analyzed for response to WNT974. The ascites samples that showed a decrease in ATP concentration after treatment demonstrated no difference from the untreated cells in percent viability with trypan blue staining. Flow cytometry demonstrated fewer cells in the G2 phase and more in the G1 and S phases after treatment with WNT974. Combination therapy with WNT974 and carboplatin resulted in a higher percentage of samples that showed ≥30% reduction in ATP concentration than either single drug treatment. IHC analysis of Wnt pathway proteins suggests cell cycle arrest rather than cytotoxicity after WNT974 treatment. QPCR indicated that RSPO fusions are not prevalent in ovarian cancer tissues or ascites. However, higher PORCN expression correlated to sensitivity to WNT974 (P=0.0073). In conclusion, WNT974 produces cytostatic effects in patient ascites cells with primary ovarian cancer through inhibition of the Wnt/β-catenin pathway. The combination of WNT974 and carboplatin induces cytotoxicity plus cell cycle arrest in a higher percentage of ascites samples than with single drug treatment. RSPO fusions do not contribute to WNT974 sensitivity; however, higher PORCN expression indicates increased WNT974 sensitivity.
Collapse
Affiliation(s)
- Jonathan D Boone
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rebecca C Arend
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Sara J Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Scott A Gilchrist
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Denise K Oelschlager
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William E Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gerald McGwin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Abhishek Gangrade
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Michael Straughn
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donald J Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
35
|
Liu D, Mai K, Zhang Y, Xu W, Ai Q. GSK-3b participates in the regulation of hepatic lipid deposition in large yellow croaker (Larmichthys crocea). FISH PHYSIOLOGY AND BIOCHEMISTRY 2016; 42:379-388. [PMID: 26483261 DOI: 10.1007/s10695-015-0145-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 10/13/2015] [Indexed: 06/05/2023]
Abstract
In this study, the participation of glycogen synthase kinase-3β (GSK-3β) in the lipid deposition was investigated in the liver of large yellow croaker (Larmichthys crocea) by LiCl treatment. It was found that the expression of GSK-3β and peroxisome proliferator-activated receptor-γ (PPARγ) was inhibited, but the expression of β-catenin was induced by LiCl treatment. Furthermore, the gene expression and activity of fatty acid synthetase (FAS) and lipoprotein lipase (LPL) in the liver was inhibited by LiCl treatment. The content of total cholesterol (TC), triglyceride (TG), and non-estesterified fatty acid in the liver, as well as TC, TG, and low-density lipoprotein cholesterol in plasma, was decreased by LiCl treatment. However, high-density lipoprotein cholesterol in plasma was increased, and the number of lipid droplets in the liver was decreased by LiCl treatment. The results indicate that GSK-3β/β-catenin may participate in regulating LPL and FAS through PPARγ in the liver of large yellow croaker, which will lead to the inhibition of hepatic lipid deposition.
Collapse
|
36
|
Liu D, Mai K, Zhang Y, Xu W, Ai Q. Wnt/β-catenin signaling participates in the regulation of lipogenesis in the liver of juvenile turbot (Scophthalmus maximus L.). Comp Biochem Physiol B Biochem Mol Biol 2016; 191:155-62. [DOI: 10.1016/j.cbpb.2015.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 10/29/2015] [Accepted: 11/01/2015] [Indexed: 01/20/2023]
|
37
|
Xiao YF, Yong X, Tang B, Qin Y, Zhang JW, Zhang D, Xie R, Yang SM. Notch and Wnt signaling pathway in cancer: Crucial role and potential therapeutic targets (Review). Int J Oncol 2015; 48:437-49. [PMID: 26648421 DOI: 10.3892/ijo.2015.3280] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 10/19/2015] [Indexed: 11/05/2022] Open
Abstract
There is no radical cure for all cancer types. The most frequently used therapies are surgical treatment, radiotherapy and chemotherapy. However, recrudescence, radiation resistance and chemotherapy resistance are the most challenging issues in clinical practice. To address these issues, they should be further studied at the molecular level, and the signaling pathways involved represent a promising avenue for this research. In the present review, we mainly discuss the components and mechanisms of activation of the Notch and Wnt signaling pathways, and we summarize the recent research efforts on these two pathways in different cancers. We also evaluate the ideal drugs that could target these two signaling pathways for cancer therapy, summarize alterations in the Notch and Wnt signaling pathways in cancer, and discuss potential signaling inhibitors as effective drugs for cancer therapy.
Collapse
Affiliation(s)
- Yu-Feng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Xin Yong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yong Qin
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Jian-Wei Zhang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Dan Zhang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Rui Xie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
38
|
Burkhalter RJ, Westfall SD, Liu Y, Stack MS. Lysophosphatidic Acid Initiates Epithelial to Mesenchymal Transition and Induces β-Catenin-mediated Transcription in Epithelial Ovarian Carcinoma. J Biol Chem 2015; 290:22143-54. [PMID: 26175151 DOI: 10.1074/jbc.m115.641092] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Indexed: 11/06/2022] Open
Abstract
During tumor progression, epithelial ovarian cancer (EOC) cells undergo epithelial-to-mesenchymal transition (EMT), which influences metastatic success. Mutation-dependent activation of Wnt/β-catenin signaling has been implicated in gain of mesenchymal phenotype and loss of differentiation in several solid tumors; however, similar mutations are rare in most EOC histotypes. Nevertheless, evidence for activated Wnt/β-catenin signaling in EOC has been reported, and immunohistochemical analysis of human EOC tumors demonstrates nuclear staining in all histotypes. This study addresses the hypothesis that the bioactive lipid lysophosphatidic acid (LPA), prevalent in the EOC microenvironment, functions to regulate EMT in EOC. Our results demonstrate that LPA induces loss of junctional β-catenin, stimulates clustering of β1 integrins, and enhances the conformationally active population of surface β1 integrins. Furthermore, LPA treatment initiates nuclear translocation of β-catenin and transcriptional activation of Wnt/β-catenin target genes resulting in gain of mesenchymal marker expression. Together these data suggest that LPA initiates EMT in ovarian tumors through β1-integrin-dependent activation of Wnt/β-catenin signaling, providing a novel mechanism for mutation-independent activation of this pathway in EOC progression.
Collapse
Affiliation(s)
- Rebecca J Burkhalter
- From the Departments of Medical Pharmacology and Physiology and the Harper Cancer Research Institute
| | - Suzanne D Westfall
- Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri 65212 and
| | - Yueying Liu
- the Harper Cancer Research Institute, Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana 46617
| | - M Sharon Stack
- the Harper Cancer Research Institute, Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana 46617
| |
Collapse
|
39
|
Bobbs A, Gellerman K, Hallas WM, Joseph S, Yang C, Kurkewich J, Cowden Dahl KD. ARID3B Directly Regulates Ovarian Cancer Promoting Genes. PLoS One 2015; 10:e0131961. [PMID: 26121572 PMCID: PMC4486168 DOI: 10.1371/journal.pone.0131961] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 06/08/2015] [Indexed: 01/22/2023] Open
Abstract
The DNA-binding protein AT-Rich Interactive Domain 3B (ARID3B) is elevated in ovarian cancer and increases tumor growth in a xenograft model of ovarian cancer. However, relatively little is known about ARID3B's function. In this study we perform the first genome wide screen for ARID3B direct target genes and ARID3B regulated pathways. We identified and confirmed numerous ARID3B target genes by chromatin immunoprecipitation (ChIP) followed by microarray and quantitative RT-PCR. Using motif-finding algorithms, we characterized a binding site for ARID3B, which is similar to the previously known site for the ARID3B paralogue ARID3A. Functionality of this predicted site was demonstrated by ChIP analysis. We next demonstrated that ARID3B induces expression of its targets in ovarian cancer cell lines. We validated that ARID3B binds to an epidermal growth factor receptor (EGFR) enhancer and increases mRNA expression. ARID3B also binds to the promoter of Wnt5A and its receptor FZD5. FZD5 is highly expressed in ovarian cancer cell lines, and is upregulated by exogenous ARID3B. Both ARID3B and FZD5 expression increase adhesion to extracellular matrix (ECM) components including collagen IV, fibronectin and vitronectin. ARID3B-increased adhesion to collagens II and IV require FZD5. This study directly demonstrates that ARID3B binds target genes in a sequence-specific manner, resulting in increased gene expression. Furthermore, our data indicate that ARID3B regulation of direct target genes in the Wnt pathway promotes adhesion of ovarian cancer cells.
Collapse
Affiliation(s)
- Alexander Bobbs
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, Indiana, United States of America
- Harper Cancer Research Institute, South Bend, Indiana, United States of America
| | - Katrina Gellerman
- Harper Cancer Research Institute, South Bend, Indiana, United States of America
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - William Morgan Hallas
- Harper Cancer Research Institute, South Bend, Indiana, United States of America
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Stancy Joseph
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, Indiana, United States of America
- Harper Cancer Research Institute, South Bend, Indiana, United States of America
| | - Chao Yang
- Harper Cancer Research Institute, South Bend, Indiana, United States of America
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Jeffrey Kurkewich
- Harper Cancer Research Institute, South Bend, Indiana, United States of America
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Karen D. Cowden Dahl
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, Indiana, United States of America
- Harper Cancer Research Institute, South Bend, Indiana, United States of America
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
40
|
Giannakouros P, Comamala M, Matte I, Rancourt C, Piché A. MUC16 mucin (CA125) regulates the formation of multicellular aggregates by altering β-catenin signaling. Am J Cancer Res 2014; 5:219-230. [PMID: 25628932 PMCID: PMC4300699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 11/20/2014] [Indexed: 06/04/2023] Open
Abstract
After shedding from the primary tumor site, ovarian cancer cells form three-dimensional multicellular aggregates that serve as vehicle for cancer cell dissemination in the peritoneal cavity. MUC16 mucin (CA125) is aberrantly expressed by most advanced serous ovarian cancers and can promote proliferation, migration and metastasis. MUC16 associates with E-cadherin and β-catenin, two proteins involved in regulation of cell adhesion and the formation of multicellular aggregates. However, the role of MUC16 in the formation of multicellular aggregates remains to be defined. Here, we show that MUC16 alters E-cadherin cellular localization and expression. Consistent with this, MUC16 knockdown inhibited the formation of multicellular aggregates and, conversely, forced expression of MUC16 C-terminal domain (CTD) enhanced the formation of multicellular aggregates. MUC16 knockdown induces β-catenin relocation from the cell membrane to the cytoplasm, decreases its expression by increasing degradation and decreases β-catenin target gene expression. MUC16 CTD inhibits GSK-3β-mediated phosphorylation and degradation of β-catenin, leading to increased β-catenin levels. Importantly, knockdown of β-catenin inhibited multicellular aggregation. These findings indicate that MUC16 promotes the formation of multicellular aggregates by inhibiting β-catenin degradation.
Collapse
Affiliation(s)
- Panagiota Giannakouros
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke 3001, 12 ième Avenue Nord, Sherbrooke, Canada J1H 5N4
| | - Marina Comamala
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke 3001, 12 ième Avenue Nord, Sherbrooke, Canada J1H 5N4
| | - Isabelle Matte
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke 3001, 12 ième Avenue Nord, Sherbrooke, Canada J1H 5N4
| | - Claudine Rancourt
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke 3001, 12 ième Avenue Nord, Sherbrooke, Canada J1H 5N4
| | - Alain Piché
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke 3001, 12 ième Avenue Nord, Sherbrooke, Canada J1H 5N4
| |
Collapse
|
41
|
Desjardins M, Xie J, Gurler H, Muralidhar GG, Sacks JD, Burdette JE, Barbolina MV. Versican regulates metastasis of epithelial ovarian carcinoma cells and spheroids. J Ovarian Res 2014; 7:70. [PMID: 24999371 PMCID: PMC4081460 DOI: 10.1186/1757-2215-7-70] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 06/19/2014] [Indexed: 01/08/2023] Open
Abstract
Background Epithelial ovarian carcinoma is a deadly disease characterized by overt peritoneal metastasis. Individual cells and multicellular aggregates, or spheroids, seed these metastases, both commonly found in ascites. Mechanisms that foster spheroid attachment to the peritoneal tissues preceding formation of secondary lesions are largely unknown. Methods Cell culture models of SKOV-3, OVCAR3, OVCAR4, Caov-3, IGROV-1, and A2780 were used. In this report the role of versican was examined in adhesion of EOC spheroids and cells to peritoneal mesothelial cell monolayers in vitro as well as in formation of peritoneal tumors using an in vivo xenograft mouse model. Results The data demonstrate that versican is instrumental in facilitating cell and spheroid adhesion to the mesothelial cell monolayers, as its reduction with specific shRNAs led to decreased adhesion. Furthermore, spheroids with reduced expression of versican failed to disaggregate to complete monolayers when seeded atop monolayers of peritoneal mesothelial cells. Failure of spheroids lacking versican to disaggregate as efficiently as controls could be attributed to a reduced cell migration that was observed in the absence of versican expression. Importantly, both spheroids and cells with reduced expression of versican demonstrated significantly impaired ability to generate peritoneal tumors when injected intraperitoneally into athymic nude mice. Conclusions Taken together these data suggest that versican regulates the development of peritoneal metastasis originating from cells and spheroids.
Collapse
Affiliation(s)
- Mark Desjardins
- Departments of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 South Wood Street, PHARM 335, Chicago, IL 60612, USA
| | - Jia Xie
- Departments of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 South Wood Street, PHARM 335, Chicago, IL 60612, USA
| | - Hilal Gurler
- Departments of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 South Wood Street, PHARM 335, Chicago, IL 60612, USA
| | - Goda G Muralidhar
- Departments of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 South Wood Street, PHARM 335, Chicago, IL 60612, USA
| | - Joelle D Sacks
- Departments of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 South Wood Street, PHARM 335, Chicago, IL 60612, USA
| | - Joanna E Burdette
- Medicinal Chemistry and Pharmacognocy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Maria V Barbolina
- Departments of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 South Wood Street, PHARM 335, Chicago, IL 60612, USA
| |
Collapse
|
42
|
Pettee KM, Dvorak KM, Nestor-Kalinoski AL, Eisenmann KM. An mDia2/ROCK signaling axis regulates invasive egress from epithelial ovarian cancer spheroids. PLoS One 2014; 9:e90371. [PMID: 24587343 PMCID: PMC3938721 DOI: 10.1371/journal.pone.0090371] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 02/03/2014] [Indexed: 12/22/2022] Open
Abstract
Multi-cellular spheroids are enriched in ascites of epithelial ovarian cancer (OvCa) patients. They represent an invasive and chemoresistant cellular population fundamental to metastatic dissemination. The molecular mechanisms triggering single cell invasive egress from spheroids remain enigmatic. mDia formins are Rho GTPase effectors that are key regulators of F-actin cytoskeletal dynamics. We hypothesized that mDia2-driven F-actin dynamics promote single cell invasive transitions in clinically relevant three-dimensional (3D) OvCa spheroids. The current study is a dissection of the contribution of the F-actin assembly factor mDia2 formin in invasive transitions and using a clinically relevant ovarian cancer spheroid model. We show that RhoA-directed mDia2 activity is required for tight spheroid organization, and enrichment of mDia2 in the invasive cellular protrusions of collagen-embedded OVCA429 spheroids. Depleting mDia2 in ES-2 spheroids enhanced invasive dissemination of single amoeboid-shaped cells. This contrasts with spheroids treated with control siRNA, where a mesenchymal invasion program predominated. Inhibition of another RhoA effector, ROCK, had no impact on ES-2 spheroid formation but dramatically inhibited spheroid invasion through induction of a highly elongated morphology. Concurrent inhibition of ROCK and mDia2 blocked single cell invasion from ES-2 spheroids more effectively than inhibition of either protein alone, indicating that invasive egress of amoeboid cells from mDia2-depleted spheroids is ROCK-dependent. Our findings indicate that multiple GTPase effectors must be suppressed in order to fully block invasive egress from ovarian cancer spheroids. Furthermore, tightly regulated interplay between ROCK and mDia2 signaling pathways dictates the invasive capacities and the type of invasion program utilized by motile spheroid-derived ovarian cancer cells. As loss of the gene encoding mDia2, DRF3, has been linked to cancer progression and metastasis, our results set the stage for understanding molecular mechanisms involved in mDia2-dependent egress of invasive cells from primary epithelial tumors.
Collapse
MESH Headings
- Actin Cytoskeleton/chemistry
- Actin Cytoskeleton/metabolism
- Carcinoma, Ovarian Epithelial
- Carrier Proteins/antagonists & inhibitors
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Cell Movement
- Female
- Formins
- Gene Expression Regulation, Neoplastic
- Humans
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/metabolism
- Neoplasms, Glandular and Epithelial/pathology
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- rho-Associated Kinases/antagonists & inhibitors
- rho-Associated Kinases/genetics
- rho-Associated Kinases/metabolism
- rhoA GTP-Binding Protein/genetics
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Krista M. Pettee
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, Ohio, United States of America
| | - Kaitlyn M. Dvorak
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, Ohio, United States of America
| | - Andrea L. Nestor-Kalinoski
- Department of Surgery, University of Toledo Health Science Campus, Toledo, Ohio, United States of America
| | - Kathryn M. Eisenmann
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, Ohio, United States of America
- * E-mail:
| |
Collapse
|
43
|
Brosseau JP, Lucier JF, Nwilati H, Thibault P, Garneau D, Gendron D, Durand M, Couture S, Lapointe E, Prinos P, Klinck R, Perreault JP, Chabot B, Abou-Elela S. Tumor microenvironment-associated modifications of alternative splicing. RNA (NEW YORK, N.Y.) 2014; 20:189-201. [PMID: 24335142 PMCID: PMC3895271 DOI: 10.1261/rna.042168.113] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Pre-mRNA alternative splicing is modified in cancer, but the origin and specificity of these changes remain unclear. Here, we probed ovarian tumors to identify cancer-associated splicing isoforms and define the mechanism by which splicing is modified in cancer cells. Using high-throughput quantitative PCR, we monitored the expression of splice variants in laser-dissected tissues from ovarian tumors. Surprisingly, changes in alternative splicing were not limited to the tumor tissues but were also found in the tumor microenvironment. Changes in the tumor-associated splicing events were found to be regulated by splicing factors that are differentially expressed in cancer tissues. Overall, ∼20% of the alternative splicing events affected by the down-regulation of the splicing factors QKI and RBFOX2 were altered in the microenvironment of ovarian tumors. Together, our results indicate that the tumor microenvironment undergoes specific changes in alternative splicing orchestrated by a limited number of splicing factors.
Collapse
Affiliation(s)
- Jean-Philippe Brosseau
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Jean-François Lucier
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Hanad Nwilati
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Philippe Thibault
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Daniel Garneau
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Daniel Gendron
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Mathieu Durand
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Sonia Couture
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Elvy Lapointe
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Panagiotis Prinos
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Roscoe Klinck
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Jean-Pierre Perreault
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Benoit Chabot
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Sherif Abou-Elela
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Corresponding authorE-mail
| |
Collapse
|
44
|
Basu M, Mukhopadhyay S, Chatterjee U, Roy SS. FGF16 promotes invasive behavior of SKOV-3 ovarian cancer cells through activation of mitogen-activated protein kinase (MAPK) signaling pathway. J Biol Chem 2014; 289:1415-28. [PMID: 24253043 PMCID: PMC3894325 DOI: 10.1074/jbc.m113.535427] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Indexed: 12/12/2022] Open
Abstract
Uncontrolled cell growth and tissue invasion define the characteristic features of cancer. Several growth factors regulate these processes by inducing specific signaling pathways. We show that FGF16, a novel factor, is expressed in human ovary, and its expression is markedly increased in ovarian tumors. This finding indicated possible involvement of FGF16 in ovarian cancer progression. We observed that FGF16 stimulates the proliferation of human ovarian adenocarcinoma cells, SKOV-3 and OAW-42. Furthermore, through the activation of FGF receptor-mediated intracellular MAPK pathway, FGF16 regulates the expression of MMP2, MMP9, SNAI1, and CDH1 and thus facilitates cellular invasion. Inhibition of FGFR as well as MAPK pathway reduces the proliferative and invasive behavior of ovarian cancer cells. Moreover, ovarian tumors with up-regulated PITX2 expression also showed activation of Wnt/β-catenin pathway that prompted us to investigate possible interaction among FGF16, PITX2, and Wnt pathway. We identified that PITX2 homeodomain transcription factor interacts with and regulates FGF16 expression. Furthermore, activation of the Wnt/β-catenin pathway induces FGF16 expression. Moreover, FGF16 promoter possesses the binding elements of PITX2 as well as T-cell factor (Wnt-responsive), in close proximity, where PITX2 and β-catenin binds to and synergistically activates the same. A detail study showed that both PITX2 and T-cell factor elements and the interaction with their binding partners are necessary for target gene expression. Taken together, our findings indicate that FGF16 in conjunction with Wnt pathway contributes to the cancer phenotype of ovarian cells and suggests that modulation of its expression in ovarian cells might be a promising therapeutic strategy for the treatment of invasive ovarian cancers.
Collapse
Affiliation(s)
- Moitri Basu
- From the Cell Biology and Physiology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India and
| | | | - Uttara Chatterjee
- Department of Pathology, Institute of Post Graduate Medical Education and Research and Seth Sukhlal Karnani Memorial Hospital, 244 AJC Bose Road, Kolkata 700020, India
| | - Sib Sankar Roy
- From the Cell Biology and Physiology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India and
| |
Collapse
|
45
|
Arend RC, Londoño-Joshi AI, Straughn JM, Buchsbaum DJ. The Wnt/β-catenin pathway in ovarian cancer: a review. Gynecol Oncol 2013; 131:772-9. [PMID: 24125749 DOI: 10.1016/j.ygyno.2013.09.034] [Citation(s) in RCA: 361] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/27/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Ovarian cancer is the deadliest gynecologic malignancy and the fifth leading cause of death from cancer in women in the U.S. Since overall survival remains poor, there is a need for new therapeutic paradigms. This paper will review the Wnt/β-catenin pathway as it relates to epithelial ovarian cancer, specifically its role in chemoresistance and its potential role as a target for chemosensitization. METHODS A PubMed search was performed for articles published pertaining to Wnt/β-catenin pathway specific to ovarian cancer. Wnt/β-catenin signaling pathways play an active role in cancer stem cells (CSCs) and carcinogenesis of all ovarian cancer subtypes. Studies also have shown that ovarian CSCs are involved in chemoresistance, metastasis, and tumor recurrence. RESULTS Wnt/β-catenin target genes regulate cell proliferation and apoptosis, thereby mediating cancer initiation and progression. The Wnt/β-catenin pathway is one of the major signaling pathways thought to be involved in epithelial-to-mesenchymal transition (EMT). Alterations affecting Wnt pathway proteins on the cell membrane, in the cytoplasm, and in the nucleus have been shown to play important roles in the tumorigenesis of ovarian cancer. CONCLUSIONS Wnt signaling is activated in epithelial ovarian cancer. Given the role of the Wnt/β-catenin pathway in carcinogenesis, more pre-clinical studies are warranted to further investigate other Wnt inhibitors in ovarian cancer. The Wnt pathway should also be investigated as a potential target in the development of new drugs for ovarian cancer as a single agent and in combination with chemotherapy or other targeted agents.
Collapse
Affiliation(s)
- Rebecca C Arend
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, USA.
| | | | | | | |
Collapse
|
46
|
Longuespée R, Boyon C, Desmons A, Vinatier D, Leblanc E, Farré I, Wisztorski M, Ly K, D'Anjou F, Day R, Fournier I, Salzet M. Ovarian cancer molecular pathology. Cancer Metastasis Rev 2013; 31:713-32. [PMID: 22729278 DOI: 10.1007/s10555-012-9383-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ovarian cancer (OVC) is the fourth leading cause of cancer mortality among women in Europe and the United States. Its early detection is difficult due to the lack of specificity of clinical symptoms. Unfortunately, late diagnosis is a major contributor to the poor survival rates for OVC, which can be attributed to the lack of specific sets of markers. Aside from patients sharing a strong family history of ovarian and breast cancer, including the BRCA1 and BRCA2 tumor suppressor genes mutations, the most used biomarker is the Cancer-antigen 125 (CA-125). CA-125 has a sensitivity of 80 % and a specificity of 97 % in epithelial cancer (stage III or IV). However, its sensitivity is 30 % in stage I cancer, as its increase is linked to several physiological phenomena and benign situations. CA-125 is particularly useful for at-risk population diagnosis and to assess response to treatment. It is clear that alone, CA-125 is inadequate as a biomarker for OVC diagnosis. There is an unmet need to identify additional biomarkers. Novel and more sensitive proteomic strategies such as MALDI mass spectrometry imaging studies are well suited to identify better markers for both diagnosis and prognosis. In the present review, we will focus on such proteomic strategies in regards to OVC signaling pathways, OVC development and escape from the immune response.
Collapse
Affiliation(s)
- Rémi Longuespée
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée, Université Nord de France, EA 4550, Université de Lille 1, Cité Scientifique, 59650 Villeneuve D'Ascq, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kwon MJ, Shin YK. Regulation of ovarian cancer stem cells or tumor-initiating cells. Int J Mol Sci 2013; 14:6624-48. [PMID: 23528891 PMCID: PMC3645658 DOI: 10.3390/ijms14046624] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 03/08/2013] [Accepted: 03/12/2013] [Indexed: 12/18/2022] Open
Abstract
Cancer stem cells or tumor-initiating cells (CSC/TICs), which can undergo self-renewal and differentiation, are thought to play critical roles in tumorigenesis, therapy resistance, tumor recurrence and metastasis. Tumor recurrence and chemoresistance are major causes of poor survival rates of ovarian cancer patients, which may be due in part to the existence of CSC/TICs. Therefore, elucidating the molecular mechanisms responsible for the ovarian CSC/TICs is required to develop a cure for this malignancy. Recent studies have indicated that the properties of CSC/TICs can be regulated by microRNAs, genes and signaling pathways which also function in normal stem cells. Moreover, emerging evidence suggests that the tumor microenvironments surrounding CSC/TICs are crucial for the maintenance of these cells. Similarly, efforts are now being made to unravel the mechanism involved in the regulation of ovarian CSC/TICs, although much work is still needed. This review considers recent advances in identifying the genes and pathways involved in the regulation of ovarian CSC/TICs. Furthermore, current approaches targeting ovarian CSC/TICs are described. Targeting both CSC/TICs and bulk tumor cells is suggested as a more effective approach to eliminating ovarian tumors. Better understanding of the regulation of ovarian CSC/TICs might facilitate the development of improved therapeutic strategies for recurrent ovarian cancer.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 702-701, Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 702-701, Korea
- Authors to whom correspondence should be addressed: E-Mails: (M.J.K.); (Y.K.S.); Tel.: +82-53-950-8581 (M.J.K.); +82-2-880-9126 (Y.K.S.); Fax: +82-53-950-8557 (M.J.K.); +82-2-883-9126 (Y.K.S.)
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
- Advanced Institutes of Convergence Technology, Suwon, Gyeonggi-do 443-270, Korea
- Authors to whom correspondence should be addressed: E-Mails: (M.J.K.); (Y.K.S.); Tel.: +82-53-950-8581 (M.J.K.); +82-2-880-9126 (Y.K.S.); Fax: +82-53-950-8557 (M.J.K.); +82-2-883-9126 (Y.K.S.)
| |
Collapse
|
48
|
Basu M, Roy SS. Wnt/β-catenin pathway is regulated by PITX2 homeodomain protein and thus contributes to the proliferation of human ovarian adenocarcinoma cell, SKOV-3. J Biol Chem 2013; 288:4355-67. [PMID: 23250740 PMCID: PMC3567686 DOI: 10.1074/jbc.m112.409102] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 11/26/2012] [Indexed: 01/22/2023] Open
Abstract
Pituitary homeobox-2 (PITX2) plays a substantial role in the development of pituitary, heart, and brain. Although the role of PITX2 isoforms in embryonic development has been extensively studied, its possible involvement in regulating the Wnt signaling pathway has not been reported. Because the Wnt pathway is strongly involved in ovarian development and cancer, we focused on the possible association between PITX2 and Wnt pathway in ovarian carcinoma cells. Remarkably, we found that PITX2 interacts and regulates WNT2/5A/9A/6/2B genes of the canonical, noncanonical, or other pathways in the human ovarian cancer cell SKOV-3. Chromatin immunoprecipitation and promoter-reporter assays further indicated the significant association of PITX2 with WNT2 and WNT5A promoters. Detailed study further reveals that the PITX2 isoform specifically activates the canonical Wnt signaling pathway either directly or through Wnt ligands. Thus, the activated Wnt pathway subsequently enhances cell proliferation. Moreover, we found the activation of Wnt pathway reduces the expression of different FZD receptors that limit further Wnt activation, demonstrating the existence of an auto-regulatory feedback loop. In contrast, PITX2 could not activate the noncanonical pathway as the Wnt5A-specific ROR2 receptor does not express in SKOV-3 cells. Collectively, our findings demonstrated that, despite being a target of the canonical Wnt signaling pathway, PITX2 itself induces the same, thus leading to the activation of the cell cycle regulating genes as well as the proliferation of SKOV-3 cells. Collectively, we highlighted that the PITX2 and Wnt pathway exerts a positive feedback regulation, whereas frizzled receptors generate a negative feedback in this pathway. Our findings will help to understand the molecular mechanism of proliferation in ovarian cancer cells.
Collapse
Affiliation(s)
- Moitri Basu
- From the Cell Biology and Physiology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja Subodh Chandra Mullick Road, Kolkata 700032, India
| | - Sib Sankar Roy
- From the Cell Biology and Physiology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja Subodh Chandra Mullick Road, Kolkata 700032, India
| |
Collapse
|
49
|
Ford CE, Jary E, Ma SSQ, Nixdorf S, Heinzelmann-Schwarz VA, Ward RL. The Wnt gatekeeper SFRP4 modulates EMT, cell migration and downstream Wnt signalling in serous ovarian cancer cells. PLoS One 2013; 8:e54362. [PMID: 23326605 PMCID: PMC3543420 DOI: 10.1371/journal.pone.0054362] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 12/11/2012] [Indexed: 12/31/2022] Open
Abstract
Aberrant Wnt signalling is implicated in numerous human cancers, and understanding the effects of modulation of pathway members may lead to the development of novel therapeutics. Expression of secreted frizzled related protein 4 (SFRP4), an extracellular modulator of the Wnt signalling pathway, is progressively lost in more aggressive ovarian cancer phenotypes. Here we show that recombinant SFRP4 (rSFRP4) treatment of a serous ovarian cancer cell line results in inhibition of β-catenin dependent Wnt signalling as measured by TOP/FOP Wnt reporter assay and decreased transcription of Wnt target genes, Axin2, CyclinD1 and Myc. In addition, rSFRP4 treatment significantly increased the ability of ovarian cancer cells to adhere to collagen and fibronectin, and decreased their ability to migrate across an inflicted wound. We conclude that these changes in cell behaviour may be mediated via mesenchymal to epithelial transition (MET), as rSFRP4 treatment also resulted in increased expression of the epithelial marker E-cadherin, and reduced expression of Vimentin and Twist. Combined, these results indicate that modulation of a single upstream gatekeeper of Wnt signalling can have effects on downstream Wnt signalling and ovarian cancer cell behaviour, as mediated through epithelial to mesenchymal plasticity (EMP). This raises the possibility that SFRP4 may be used both diagnostically and therapeutically in epithelial ovarian cancer.
Collapse
Affiliation(s)
- Caroline E Ford
- Wnt Signalling & Metastasis Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia.
| | | | | | | | | | | |
Collapse
|
50
|
Loessner D, Little JP, Pettet GJ, Hutmacher DW. A multiscale road map of cancer spheroids – incorporating experimental and mathematical modelling to understand cancer progression. J Cell Sci 2013; 126:2761-71. [DOI: 10.1242/jcs.123836] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Computational models represent a highly suitable framework, not only for testing biological hypotheses and generating new ones but also for optimising experimental strategies. As one surveys the literature devoted to cancer modelling, it is obvious that immense progress has been made in applying simulation techniques to the study of cancer biology, although the full impact has yet to be realised. For example, there are excellent models to describe cancer incidence rates or factors for early disease detection, but these predictions are unable to explain the functional and molecular changes that are associated with tumour progression. In addition, it is crucial that interactions between mechanical effects, and intracellular and intercellular signalling are incorporated in order to understand cancer growth, its interaction with the extracellular microenvironment and invasion of secondary sites. There is a compelling need to tailor new, physiologically relevant in silico models that are specialised for particular types of cancer, such as ovarian cancer owing to its unique route of metastasis, which are capable of investigating anti-cancer therapies, and generating both qualitative and quantitative predictions. This Commentary will focus on how computational simulation approaches can advance our understanding of ovarian cancer progression and treatment, in particular, with the help of multicellular cancer spheroids, and thus, can inform biological hypothesis and experimental design.
Collapse
|