1
|
Shichi Y, Gomi F, Ueda Y, Nonaka K, Hasegawa F, Hasegawa Y, Hinata N, Yoshimura H, Yamamoto M, Takahashi K, Arai T, Ishiwata T. Multiple cystic sphere formation from PK-8 cells in three-dimensional culture. Biochem Biophys Rep 2022; 32:101339. [PMID: 36105614 PMCID: PMC9464880 DOI: 10.1016/j.bbrep.2022.101339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022] Open
Abstract
Three-dimensional (3D) culture of cancer cells mimics the in vivo environment. Recently, we reported that pancreatic ductal adenocarcinoma (PDAC) cell lines with epithelial and mesenchymal features formed differently shaped spheres in 3D culture. However, only PK-8 cells, the epithelial PDAC cell line with the highest E-cadherin expression among the eight PDAC cell lines, formed multiple cystic spheres in 3D culture. Optical coherence tomography revealed interconnected cysts inside the spheres. A weak inter-cellular adhesion, individual cell degeneration, necrosis, and secretory granules in the cytoplasm were observed in the PK-8 spheres using electron microscopy. The expression of MUC1, MUC5AC, and amylase was increased in PK-8 cells in the 3D culture compared with that in 2D culture. These findings suggest that highly E-cadherin-expressing epithelial PK-8 cells form multiple cystic spheres, which may be promoted by enhanced mucin and amylase synthesis in 3D culture. PK-8 cells highly express E-cadherin in multiple cystic spheres in 3D culture. The cysts within the PK-8 spheres are interconnected. Individual cell degeneration and necrosis was observed in PK-8 spheres. MUC1, MUC5, and amylase levels increased in PK-8 spheres.
Collapse
|
2
|
Xiao RR, Jin L, Xie N, Luo P, Gao W, Tu P, Ai X. Establishment and large-scale validation of a three-dimensional tumor model on an array chip for anticancer drug evaluation. Front Pharmacol 2022; 13:1032975. [PMID: 36313330 PMCID: PMC9596801 DOI: 10.3389/fphar.2022.1032975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/28/2022] [Indexed: 11/29/2022] Open
Abstract
Two-dimensional (2D) tumor model has always poorly predicted drug response of animal model due to the lack of recapitulation of tumor microenvironment. Establishing a biomimetic, controllable, and cost-effective three-dimensional (3D) model and large-scale validation of its in vivo predictivity has shown promise in bridging the gap between the 2D tumor model and animal model. Here, we established a matrigel-based 3D micro-tumor model on an array chip for large-scale anticancer drug evaluation. Compared with the 2D tumor model, the 3D tumor model on the chip showed spheroid morphology, slower proliferation kinetics, and comparable reproducibility. Next, the results of the chemotherapeutic evaluation from 18 drugs against 27 cancer cell lines showed 17.6% of drug resistance on the 3D tumor model. Moreover, the evaluation results of targeted drugs showed expected sensitivity and higher specificity on the 3D tumor model compared with the 2D model. Finally, the evaluation results on the 3D tumor model were more consistent with the in vivo cell-derived xenograft model, and excluded 95% false-positive results from the 2D model. Overall, the matrigel-based 3D micro-tumor model on the array chip provides a promising tool to accelerate anticancer drug discovery.
Collapse
Affiliation(s)
- Rong-Rong Xiao
- R&D Department, Beijing Daxiang Biotech Co., Ltd., Beijing, China
| | - Lei Jin
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Nan Xie
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Piaopiao Luo
- R&D Department, Beijing Daxiang Biotech Co., Ltd., Beijing, China
| | - Wenjie Gao
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoni Ai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- *Correspondence: Xiaoni Ai,
| |
Collapse
|
3
|
Sullivan L, Pacheco RR, Kmeid M, Chen A, Lee H. Tumor Stroma Ratio and Its Significance in Locally Advanced Colorectal Cancer. Curr Oncol 2022; 29:3232-3241. [PMID: 35621653 PMCID: PMC9139914 DOI: 10.3390/curroncol29050263] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 11/16/2022] Open
Abstract
Colorectal cancer is the third leading cause of cancer-related death, and its incidence is rising in the younger patient population. In the past decade, research has unveiled several processes (underlying tumorigenesis, many of which involve interactions between tumor cells and the surrounding tissue or tumor microenvironment (TME). Interactions between components of the TME are mediated at a sub-microscopic level. However, the endpoint of those interactions results in morphologic changes which can be readily assessed at microscopic examination of biopsy and resection specimens. Among these morphologic changes, alteration to the tumor stroma is a new, important determinant of colorectal cancer progression. Different methodologies to estimate the proportion of tumor stroma relative to tumor cells, or tumor stroma ratio (TSR), have been developed. Subsequent validation has supported the prognostic value, reproducibility and feasibility of TSR in various subgroups of colorectal cancer. In this manuscript, we review the literature surrounding TME in colorectal cancer, with a focus on tumor stroma ratio.
Collapse
|
4
|
He MY, Halford MM, Liu R, Roy JP, Grant ZL, Coultas L, Thio N, Gilan O, Chan YC, Dawson MA, Achen MG, Stacker SA. Three-dimensional CRISPR screening reveals epigenetic interaction with anti-angiogenic therapy. Commun Biol 2021; 4:878. [PMID: 34267311 PMCID: PMC8282794 DOI: 10.1038/s42003-021-02397-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis underlies development, physiology and pathogenesis of cancer, eye and cardiovascular diseases. Inhibiting aberrant angiogenesis using anti-angiogenic therapy (AAT) has been successful in the clinical treatment of cancer and eye diseases. However, resistance to AAT inevitably occurs and its molecular basis remains poorly understood. Here, we uncover molecular modifiers of the blood endothelial cell (EC) response to a widely used AAT bevacizumab by performing a pooled genetic screen using three-dimensional microcarrier-based cell culture and CRISPR–Cas9. Functional inhibition of the epigenetic reader BET family of proteins BRD2/3/4 shows unexpected mitigating effects on EC survival and/or proliferation upon VEGFA blockade. Moreover, transcriptomic and pathway analyses reveal an interaction between epigenetic regulation and anti-angiogenesis, which may affect chromosomal structure and activity in ECs via the cell cycle regulator CDC25B phosphatase. Collectively, our findings provide insight into epigenetic regulation of the EC response to VEGFA blockade and may facilitate development of quality biomarkers and strategies for overcoming resistance to AAT. Through three-dimensional CRISPR screening, He et al. report that functional inhibition of BET family of proteins BRD2/3/4 shows mitigating effects on blood endothelial cell (EC) survival and/or proliferation upon VEGFA blockade. An interaction between epigenetic regulation and anti-angiogenesis, which may affect chromosomal structure and activity in ECs through CDC25B phosphatase, is potentially involved with EC resistance to anti-angiogenic therapy.
Collapse
Affiliation(s)
- Michael Y He
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michael M Halford
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Ruofei Liu
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - James P Roy
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Zoe L Grant
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.,Gladstone Institutes, San Francisco, CA, USA
| | - Leigh Coultas
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Niko Thio
- Bioinformatics Core, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Omer Gilan
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Yih-Chih Chan
- Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mark A Dawson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Centre for Cancer Research, The University of Melbourne, Parkville, VIC, Australia.,Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Marc G Achen
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.,St Vincent's Institute of Medical Research, Melbourne, VIC, Australia
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia. .,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
5
|
Morphofunctional analysis of human pancreatic cancer cell lines in 2- and 3-dimensional cultures. Sci Rep 2021; 11:6775. [PMID: 33762591 PMCID: PMC7990961 DOI: 10.1038/s41598-021-86028-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/10/2021] [Indexed: 12/18/2022] Open
Abstract
Genetic, transcriptional, and morphological differences have been reported in pancreatic ductal adenocarcinoma (PDAC) cases. We recently found that epithelial or mesenchymal features were enhanced in three-dimensional (3D) cultures compared to two-dimensional (2D) cultures. In this study, we examined the differences in the morphological and functional characteristics of eight PDAC cell lines in 2D and 3D cultures. Most PDAC cells showed similar pleomorphic morphologies in 2D culture. Under 3D culture, PDAC cells with high E-cadherin and low vimentin expression levels (epithelial) formed small round spheres encircled with flat lining cells, whereas those with high vimentin and low E-cadherin expression levels (mesenchymal) formed large grape-like spheres without lining cells and were highly proliferative. In 3D culture, gemcitabine was more effective for the spheres formed by PDAC cells with epithelial features, while abraxane was more effective on those with mesenchymal features. The expression levels of drug transporters were highest PDAC cells with high vimentin expression levels. These findings indicate that PDAC cells possess various levels of epithelial and mesenchymal characteristics. The 3D-culture method is useful for investigating the diversity of PDAC cell lines and may play important roles in the development of personalized early diagnostic methods and anticancer drugs for PDAC.
Collapse
|
6
|
Li Q, Zhou X, Fang Z, Pan Z. Effect of STC2 gene silencing on colorectal cancer cells. Mol Med Rep 2019; 20:977-984. [PMID: 31173256 PMCID: PMC6625197 DOI: 10.3892/mmr.2019.10332] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
Stanniocalcin 2 (STC2), a secretory glycoprotein hormone, regulates many biological processes including cell proliferation, apoptosis, tumorigenesis and atherosclerosis. However, the effect of STC2 on proliferation, migration and epithelial‑mesenchymal transition (EMT) progression in human colorectal cancer (CRC) cells remains poorly understood. The expression level of STC2 was determined by quantitative real‑time polymerase chain reaction (qPCR) and western blot analysis. Cell Counting Kit‑8 (CCK‑8) was used to detect the viability of SW480 cells. The invasion and migration of cells were identified by wound healing and Transwell assays. The mRNA and protein expression levels of β‑catenin, matrix metalloproteinase (MMP)‑2, MMP‑9, E‑cadherin and vimentin were assessed by qPCR and western blot analysis. In the present study, it was demonstrated that STC2 was highly expressed in the CRC cell lines. After silencing of STC2, the cell viability, migration and invasion were significantly reduced. Silencing of STC2 in the CRC Sw480 cells increased the expression of E‑cadherin and decreased the expression of vimentin, MMP‑2 and MMP‑9, compared to those in the normal and empty vector group. Furthermore, the expression of β‑catenin in the STC2 gene silenced group was suppressed, and the expression of β‑catenin was reversed by Wnt activator, SB216763. These results demonstrated that STC2 participates in the development and progression of CRC by promoting CRC cell proliferation, survival and migration and activating the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Qianyuan Li
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiukou Zhou
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhengyu Fang
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhiyun Pan
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
7
|
Enhanced morphological and functional differences of pancreatic cancer with epithelial or mesenchymal characteristics in 3D culture. Sci Rep 2019; 9:10871. [PMID: 31350453 PMCID: PMC6659675 DOI: 10.1038/s41598-019-47416-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer, composed of heterogeneous cancer cells, alters epithelial to mesenchymal features during growth and metastasis. In this study, we aimed to characterize pancreatic ductal adenocarcinoma (PDAC) cells showing epithelial or mesenchymal features in 3D culture. In 3D culture, PK-1 cells had high E-cadherin and low vimentin expression and exhibited a round-like appearance encircled by flat cells. PANC-1 cells had high vimentin and low E-cadherin expression and formed grape-like spheres. PK-1 cells had secretary granules and many microvilli, desmosomes, and adherens junctions, while PANC-1 cells had few microvilli, adherens junction, and no desmosomes. Cytokeratin 7, trypsin, CA19-9, and E-cadherin were highly expressed in PK-1 cells but not in PANC-1 cells. Ki-67 was diffusely expressed in PANC-1 spheres but was restricted to the peripheral flat cells of PK-1 spheres. PANC-1 and PK-1 cells were positive for transforming growth factor (TGF) β receptor II and phosphorylated smad2/3, but PK-1 cells were smad4 negative. Taken together, 3D culture enhanced morphofunctional differences of PDAC cells showing epithelial or mesenchymal characteristics, and epithelial phenotype maintenance may be due to the ineffectiveness of the TGF- β pathway. Clarification of heterogeneity using 3D culture may be useful for development of individualized diagnostic and therapeutic methods in patients with PDAC.
Collapse
|
8
|
Chen B, Zeng X, He Y, Wang X, Liang Z, Liu J, Zhang P, Zhu H, Xu N, Liang S. STC2 promotes the epithelial-mesenchymal transition of colorectal cancer cells through AKT-ERK signaling pathways. Oncotarget 2018; 7:71400-71416. [PMID: 27662663 PMCID: PMC5342087 DOI: 10.18632/oncotarget.12147] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 09/13/2016] [Indexed: 02/05/2023] Open
Abstract
The STC2 protein involves in carcinogenesis and progression of many cancers. It remains unclear how STC2 regulates the epithelial-mesenchymal transition (EMT) process and colorectal cancer (CRC) development. Here we systematically investigated STC2-activated early occurrence of EMT and CRC cell migration in vitro, clinical associations of STC2 with CRC development and patient survival. The secretion and expression level of STC2 were both greatly increased in EMT cells and CRC cells compared with the normal epithelial NCM460 cells. And the conditioned media from EMT cells stimulated epithelia and colon cancer cells to obtain EMT characteristics. STC2 overexpression promoted CRC cell growth and cell migration in vitro, and STC2 enhanced tumor growth in a mouse CRC-xenograft model. Corresponding to EMT marker expression changes, several critical signaling pathway molecules including pERK, pAKT, PI3K and Ras were remarkably increased either in NCM460 cells transfected with STC2 plasmids or in cells induced with exogenous STC2 protein. However blocking AKT-ERK signaling pathways attenuated STC2-activated EMT process. Furthermore the elevated STC2 expressions were also confirmed in 77 clinical tumor tissues and sera from CRC patients, and the increased STC2 in tumor tissues and sera correlated with tumor pathologic stage and poor survival for CRC patients. In conclusion, STC2 promotes CRC tumorigenesis and EMT progression through activating ERK/MEK and PI3K/AKT signaling pathways. STC2 protein is also a potential tumor biomarker for CRC diagnosis and prognosis.
Collapse
Affiliation(s)
- Bing Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Xiao Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Yu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Xixi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Ziwei Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Jingjing Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Peng Zhang
- Department of Urinary Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Hongxia Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China.,Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100034, P. R. China
| | - Ningzhi Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China.,Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100034, P. R. China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| |
Collapse
|
9
|
Vincan E, Schwab RHM, Flanagan DJ, Moselen JM, Tran BM, Barker N, Phesse TJ. The Central Role of Wnt Signaling and Organoid Technology in Personalizing Anticancer Therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 153:299-319. [PMID: 29389521 DOI: 10.1016/bs.pmbts.2017.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Wnt pathway is at the heart of organoid technology, which is set to revolutionize the cancer field. We can now predetermine a patient's response to any given anticancer therapy by exposing tumor organoids established from the patient's own tumor. This cutting-edge biomedical platform translates to patients being treated with the correct drug at the correct dose from the outset, a truly personalized and precise medical approach. A high throughput drug screen on organoids also allows drugs to be tested in limitless combinations. More recently, the tumor cells that are resistant to the therapy given to a patient were selected in culture using the patient's organoids. The resistant tumor organoids were then screened empirically to identify drugs that will kill the resistant cells. This information allows diagnosis in real-time to either prevent tumor recurrence or effectively treat the recurring tumor. Furthermore, the ability to culture stem cell-derived epithelium as organoids has enabled us to begin to understand how a stem cell becomes a cancer cell or to pin-point the genetic alteration that underlies a given genetic syndrome. Here we summarize these advances and the central role of Wnt signaling, and identify the next challenges for organoid technology.
Collapse
Affiliation(s)
- Elizabeth Vincan
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia; Curtin University, Perth, WA, Australia.
| | - Renate H M Schwab
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Dustin J Flanagan
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Jean M Moselen
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Bang M Tran
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Nick Barker
- A*STAR Institute of Medical Biology, Singapore
| | - Toby J Phesse
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia; European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
10
|
Schwab RHM, Amin N, Flanagan DJ, Johanson TM, Phesse TJ, Vincan E. Wnt is necessary for mesenchymal to epithelial transition in colorectal cancer cells. Dev Dyn 2017; 247:521-530. [PMID: 28560804 DOI: 10.1002/dvdy.24527] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/13/2017] [Accepted: 05/17/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Metastasis underlies most colorectal cancer mortality. Cancer cells spread through the body as single cells or small clusters of cells that have an invasive, mesenchymal, nonproliferative phenotype. At the secondary site, they revert to a proliferative "tumor constructing" epithelial phenotype to rebuild a tumor. We previously developed a unique in vitro three-dimensional model, called LIM1863-Mph, which faithfully recapitulates these reversible transitions that underpin colorectal cancer metastasis. Wnt signaling plays a key role in these transitions and is initiated by the coupling of extracellular Wnt to Frizzled (FZD). Using the LIM1863-Mph model system we demonstrated that the Wnt receptor FZD7 is necessary for mesenchymal to epithelial transition (MET). Here we investigate the role of Wnt in MET. RESULTS Wnt secretion is dependent on palmitoylation by Porcupine (PORC). A PORC inhibitor (IWP2) that prevents Wnt secretion, blocked the epithelial transition of mesenchymal LIM1863-Mph cells. Wnt gene array analysis identified several Wnts that are upregulated in epithelial compared with mesenchymal LIM1863-Mph cells, suggesting these ligands in MET. Wnt2B was the most abundant differentially expressed Wnt gene. Indeed, recombinant Wnt2B could overcome the IWP2-mediated block in epithelial transition of mesenchymal LIM1863-Mph cells. CONCLUSIONS Wnt2B co-operates with Frizzled7 to mediate MET in colorectal cancer. Developmental Dynamics 247:521-530, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Renate H M Schwab
- Molecular Oncology Laboratory, University of Melbourne and the Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Nancy Amin
- Molecular Oncology Laboratory, University of Melbourne and the Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Dustin J Flanagan
- Molecular Oncology Laboratory, University of Melbourne and the Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy M Johanson
- Molecular Oncology Laboratory, University of Melbourne and the Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Toby J Phesse
- Molecular Oncology Laboratory, University of Melbourne and the Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia.,European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff, Wales, United Kingdom
| | - Elizabeth Vincan
- Molecular Oncology Laboratory, University of Melbourne and the Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia.,School of Biomedical Sciences, Curtin University, Perth, Western Australia, Australia
| |
Collapse
|
11
|
Frizzled7: A Promising Achilles' Heel for Targeting the Wnt Receptor Complex to Treat Cancer. Cancers (Basel) 2016; 8:cancers8050050. [PMID: 27196929 PMCID: PMC4880867 DOI: 10.3390/cancers8050050] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
Frizzled7 is arguably the most studied member of the Frizzled family, which are the cognate Wnt receptors. Frizzled7 is highly conserved through evolution, from Hydra through to humans, and is expressed in diverse organisms, tissues and human disease contexts. Frizzled receptors can homo- or hetero-polymerise and associate with several co-receptors to transmit Wnt signalling. Notably, Frizzled7 can transmit signalling via multiple Wnt transduction pathways and bind to several different Wnt ligands, Frizzled receptors and co-receptors. These promiscuous binding and functional properties are thought to underlie the pivotal role Frizzled7 plays in embryonic developmental and stem cell function. Recent studies have identified that Frizzled7 is upregulated in diverse human cancers, and promotes proliferation, progression and invasion, and orchestrates cellular transitions that underscore cancer metastasis. Importantly, Frizzled7 is able to regulate Wnt signalling activity even in cancer cells which have mutations to down-stream signal transducers. In this review we discuss the various aspects of Frizzled7 signalling and function, and the implications these have for therapeutic targeting of Frizzled7 in cancer.
Collapse
|
12
|
Pulkoski-Gross A, Zheng XE, Kim D, Cathcart J, Cao J. Epithelial to Mesenchymal Transition (EMT) and Intestinal Tumorigenesis. INTESTINAL TUMORIGENESIS 2015:309-364. [DOI: 10.1007/978-3-319-19986-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
13
|
Biology and significance of circulating and disseminated tumour cells in colorectal cancer. Langenbecks Arch Surg 2012; 397:535-42. [PMID: 22350614 DOI: 10.1007/s00423-012-0917-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 01/27/2012] [Indexed: 01/15/2023]
Abstract
PURPOSE More than 130 years ago, circulating tumour cells (CTCs) and disseminated tumour cells (DTCs) have been linked to metastasis. Since then, a myriad of studies attempted to characterise and elucidate the clinical impact of CTCs/DTCs, amongst others in colorectal cancer (CRC). Due to a flood of heterogeneous findings regarding CTCs/DTCs in CRC, this review aims to describe the known facts about CTC/DTC biology and clinical impact. METHODS To identify the basic scientific literature regarding the biology and clinical impact of CTCs/DTCs in CRC, we reviewed the literature in the PubMed database. We focused on publications written in English and published until January 2012. As search terms, we used "colorectal cancer (CRC)", "colon cancer (CC)", "CTC", "DTC", "bone marrow (BM)", "lymph node (LN)", "peripheral blood (PB)", "significance" and "prognosis". RESULTS CTC detection and quantification under standardised conditions is feasible. Several studies in large patient settings have revealed prognostic impact of CTCs in CRC. CRC-derived DTC detection and analysis in BM exhibits a more heterogeneous picture but also shows clinical value. Furthermore, the presence of DTCs in LN has a strong prognostic impact in CRC. CONCLUSIONS Clinical relevance and prognostic significance of CTCs/DTCs in CRC have been clearly demonstrated in many experimental studies. The major challenge in CTC/DTC research is now to harmonise the various identification and detection approaches and consequently to conduct large prospective multi-institutional trials to verify the use of CTCs/DTCs as a valid prognostic and predictive biomarker for clinical routine.
Collapse
|
14
|
Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods 2012; 56:293-304. [PMID: 22285593 DOI: 10.1016/j.ymeth.2012.01.002] [Citation(s) in RCA: 899] [Impact Index Per Article: 69.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 01/06/2012] [Accepted: 01/10/2012] [Indexed: 12/14/2022] Open
Abstract
Exosomes are 40-100nm extracellular vesicles that are released from a multitude of cell types, and perform diverse cellular functions including intercellular communication, antigen presentation, and transfer of oncogenic proteins as well as mRNA and miRNA. Exosomes have been purified from biological fluids and in vitro cell cultures using a variety of strategies and techniques. However, all preparations invariably contain varying proportions of other membranous vesicles that co-purify with exosomes such as shed microvesicles and apoptotic blebs. Using the colorectal cancer cell line LIM1863 as a cell model, in this study we performed a comprehensive evaluation of current methods used for exosome isolation including ultracentrifugation (UC-Exos), OptiPrep™ density-based separation (DG-Exos), and immunoaffinity capture using anti-EpCAM coated magnetic beads (IAC-Exos). Notably, all isolations contained 40-100nm vesicles, and were positive for exosome markers (Alix, TSG101, HSP70) based on electron microscopy and Western blotting. We employed a proteomic approach to profile the protein composition of exosomes, and label-free spectral counting to evaluate the effectiveness of each method. Based on the number of MS/MS spectra identified for exosome markers and proteins associated with their biogenesis, trafficking, and release, we found IAC-Exos to be the most effective method to isolate exosomes. For example, Alix, TSG101, CD9 and CD81 were significantly higher (at least 2-fold) in IAC-Exos, compared to UG-Exos and DG-Exos. Application of immunoaffinity capture has enabled the identification of proteins including the ESCRT-III component VPS32C/CHMP4C, and the SNARE synaptobrevin 2 (VAMP2) in exosomes for the first time. Additionally, several cancer-related proteins were identified in IAC-Exos including various ephrins (EFNB1, EFNB2) and Eph receptors (EPHA2-8, EPHB1-4), and components involved in Wnt (CTNNB1, TNIK) and Ras (CRK, GRB2) signalling.
Collapse
|
15
|
Patel P, Chen EI. Cancer stem cells, tumor dormancy, and metastasis. Front Endocrinol (Lausanne) 2012; 3:125. [PMID: 23109929 PMCID: PMC3478572 DOI: 10.3389/fendo.2012.00125] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/02/2012] [Indexed: 12/12/2022] Open
Abstract
Tumor cells can persist undetectably for an extended period of time in primary tumors and in disseminated cancer cells. Very little is known about why and how these tumors persist for extended periods of time and then evolve to malignancy. The discovery of cancer stem cells (CSCs) in human tumors challenges our current understanding of tumor recurrence, drug resistance, and metastasis, and opens up new research directions on how cancer cells are capable of switching from dormancy to malignancy. Although overlapping molecules and pathways have been reported to regulate the stem-like phenotype of CSCs and metastasis, accumulated evidence has suggested additional clonal diversity within the stem-like cancer cell subpopulation. This review will describe the current hypothesis linking CSCs and metastasis and summarize mechanisms important for metastatic CSCs to re-initiate tumors in the secondary sites. A better understanding of CSCs' contribution to clinical tumor dormancy and metastasis will provide new therapeutic revenues to eradicate metastatic tumors and significantly reduce the mortality of cancer patients.
Collapse
Affiliation(s)
| | - Emily I. Chen
- *Correspondence: Emily I. Chen, Department of Pharmacological Sciences, Stony Brook University, BST-125, Stony Brook, NY 11794-8651, USA. e-mail:
| |
Collapse
|
16
|
Li H, Hummon AB. Imaging Mass Spectrometry of Three-Dimensional Cell Culture Systems. Anal Chem 2011; 83:8794-801. [DOI: 10.1021/ac202356g] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Haohang Li
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Amanda B. Hummon
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
17
|
Yao D, Dai C, Peng S. Mechanism of the mesenchymal-epithelial transition and its relationship with metastatic tumor formation. Mol Cancer Res 2011; 9:1608-20. [PMID: 21840933 DOI: 10.1158/1541-7786.mcr-10-0568] [Citation(s) in RCA: 331] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cancer metastasis consists of a sequential series of events, and the epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are recognized as critical events for metastasis of carcinomas. A current area of focus is the histopathological similarity between primary and metastatic tumors, and MET at sites of metastases has been postulated to be part of the process of metastatic tumor formation. Here, we summarize accumulating evidence from experimental studies that directly supports the role of MET in cancer metastasis, and we analyze the main mechanisms that regulate MET or reverse EMT in carcinomas. Given the critical role of MET in metastatic tumor formation, the potential to effectively target the MET process at sites of metastasis offers new hope for inhibiting metastatic tumor formation.
Collapse
Affiliation(s)
- Dianbo Yao
- Department of Hepatobiliary and Splenic Surgery, Shengjing Hospital of China Medical University, Heping District, Shenyang 110004, Liaoning Province, China
| | | | | |
Collapse
|
18
|
Zou J, Luo H, Zeng Q, Dong Z, Wu D, Liu L. Protein kinase CK2α is overexpressed in colorectal cancer and modulates cell proliferation and invasion via regulating EMT-related genes. J Transl Med 2011; 9:97. [PMID: 21702981 PMCID: PMC3132712 DOI: 10.1186/1479-5876-9-97] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Accepted: 06/25/2011] [Indexed: 02/08/2023] Open
Abstract
Background Protein kinase CK2 is a highly conserved, ubiquitous protein serine/threonine kinase that phosphorylates many substrates and has a global role in numerous biological and pathological processes. Overexpression of the protein kinase CK2α subunit (CK2α) has been associated with the malignant transformation of several tissues, with not nearly as much focus on the role of CK2α in colorectal cancer (CRC). The aims of this study are to investigate the function and regulatory mechanism of CK2α in CRC development. Methods Expression levels of CK2α were analyzed in 144 patients (104 with CRC and 40 with colorectal adenoma) by immunohistochemistry. Proliferation, senescence, motility and invasion assays as well as immunofluorescence staining and western blots were performed to assess the effect of CK2α in CRC. Results The immunohistochemical expression of nuclear CK2α was stronger in tumor tissues than in adenomas and normal colorectal tissues. Suppression of CK2α by small-interfering RNA or the CK2α activity inhibitor emodin inhibited proliferation of CRC cells, caused G0/G1 phase arrest, induced cell senescence, elevated the expression of p53/p21 and decreased the expression of C-myc. We also found that knockdown of CK2α suppressed cell motility and invasion. Significantly, CK2α inhibition resulted in β-catenin transactivation, decreased the expression levels of vimentin and the transcription factors snail1 and smad2/3, and increased the expression of E-cadherin, suggesting that CK2α regulates the epithelial-mesenchymal transition (EMT) process in cancer cells. Conclusions Our results indicate that CK2α plays an essential role in the development of CRC, and inhibition of CK2α may serve as a promising therapeutic strategy for human CRC.
Collapse
Affiliation(s)
- Jinjin Zou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | | | | | | | | | | |
Collapse
|
19
|
Morphological and cytoskeletal changes of pancreatic cancer cells in three-dimensional spheroidal culture. Med Mol Morphol 2011; 43:211-7. [PMID: 21267697 DOI: 10.1007/s00795-010-0497-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 01/19/2010] [Indexed: 10/18/2022]
Abstract
Three-dimensional (3D) cell cultures are expected to mimic in vivo environments. We used a NanoCulture plate to determine the spheroid-forming ability of pancreatic ductal adenocarcinoma (PDAC) cell lines and compared the morphology and expression of cytoskeletal proteins of PDAC cells to those in two-dimensional (2D) cultures. All examined PDAC cells grew as monolayers in 2D culture. PANC-1 and KLM-1 formed spheroids in 3D culture, but PK-45H and MIAPaCa-2 did not. Strong expression of F-actin was observed in the cells attached to the surface of the plate, which formed cell projections in 3D culture. F-actin was detected on the grids of the NanoCulture plate in PANC-1 cells but not in PK-45H. The levels of tubulin expression in cells were higher in 3D culture than in 2D culture. The expression level of E-cadherin mRNA in PANC-1 and KLM-1 was higher than that in PK-45H and MIAPaCa-2. In conclusion, PDAC cells showed morphological changes, spheroid formation, and alterations of cytoskeletal proteins in 3D culture. E-cadherin might be one of the key molecules involved in spheroid formation of PDAC cells. The 3D spheroidal culture system was a useful method for cell imaging with contrast-phase microscopy and confocal microscopy.
Collapse
|
20
|
Loum E, Giraud S, Bessette B, Battu S, Mathonnet M, Lautrette C. Oncogramme, a new individualized tumor response testing method: application to colon cancer. Cytotechnology 2010; 62:381-8. [PMID: 20820914 DOI: 10.1007/s10616-010-9298-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 08/06/2010] [Indexed: 11/28/2022] Open
Abstract
Colon cancer is the second leading cause of cancer-related death in industrialized countries. Many anti-cancer researches are consequently performed and individualized tumor response testing (ITRT) methods are now used to individualize patient chemotherapeutic administrations. Then, a new ITRT method, Oncogramme, was developed for colon cancer. Colon tumor fragments from different patients were dissociated and seeded in a defined culture medium. Cell preparation process as well as culture medium allowed high cell viability and a good primary culture success rate. After treatment of isolated tumoral cells by chemotherapeutics alone or in combination, cytotoxicity was determined by cell death assay allowing the Oncogramme establishment, which was validated by statistical analysis. Indeed, significant results were obtained such as different profile for each patient's cells with various drugs, and variability between patient's cells in the response to each drug. Procedure described here to obtain the Oncogramme is a new, fast and technically reliable ITRT method applied to colon cancer. For an individualized cancer treatment use, this test should be further validated by a phase I clinical trial.
Collapse
Affiliation(s)
- Elodie Loum
- Oncomedics, Faculté de Médecine, 2 rue du Docteur Marcland, 87027, Limoges, France,
| | | | | | | | | | | |
Collapse
|
21
|
Cottonham CL, Kaneko S, Xu L. miR-21 and miR-31 converge on TIAM1 to regulate migration and invasion of colon carcinoma cells. J Biol Chem 2010; 285:35293-302. [PMID: 20826792 DOI: 10.1074/jbc.m110.160069] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
TGF-β promotes cell migration and invasion, an attribute that is linked to the pro-metastasis function of this cytokine in late stage cancers. The LIM 1863 colon carcinoma organoid undergoes epithelial-mesenchymal transition (EMT) in response to TGF-β. This process is markedly accelerated by TNF-α, and we found that the levels of miR-21 and miR-31 were prominently elevated under the synergistic actions of TGF-β/TNF-α. Consistent with this, overexpression of either miR-21 or miR-31 significantly enhanced the effect of TGF-β alone on LIM 1863 morphological changes. More importantly, transwell assays demonstrated the positive effects of both miR-21 and miR-31 in TGF-β regulation of LIM 1863 motility and invasiveness. Elevated levels of miR-21 and miR-31 also enhanced motility and invasiveness of other colon carcinoma cell lines. We present compelling evidence that TIAM1, a guanidine exchange factor of the Rac GTPase, is a direct target of both miR-21 and miR-31. Indeed in LIM 1863 cells, suppression of TIAM1 is required for miR-21/miR-31 to enhance cell migration and invasion. Therefore, we have uncovered miR-21 and miR-31 as downstream effectors of TGF-β in facilitating invasion and metastasis of colon carcinoma cells.
Collapse
Affiliation(s)
- Charisa L Cottonham
- University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | |
Collapse
|
22
|
Vincan E, Flanagan DJ, Pouliot N, Brabletz T, Spaderna S. Variable FZD7 expression in colorectal cancers indicates regulation by the tumour microenvironment. Dev Dyn 2010; 239:311-7. [PMID: 19655379 DOI: 10.1002/dvdy.22045] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Recent evidence shows that a sub-population of Wnt/beta-catenin target genes is specifically induced in different tissue contexts. FZD7 is a putative Wnt/beta-catenin target gene and although it is highly expressed in well-differentiated colorectal cancer tumour cells, its expression is decreased in de-differentiated tumour cells at the invasive front despite elevated Wnt/beta-catenin signalling in this area. This variable expression of FZD7 implicates additional regulation by the microenvironment; however, this has not been investigated. To begin to elucidate the role of extracellular matrix in regulating FZD7 expression, we generated a FZD7 promoter reporter and analysed FZD7 promoter activity in colorectal cancer cells grown on different matrices. We demonstrate that the FZD7 promoter is regulated by beta-catenin in colorectal cancer cells and observed decreased promoter activity in cells grown on fibronectin but not collagen I or collagen IV. Thus, expression of FZD7 in colorectal cancer may be regulated by fibronectin in the microenvironment.
Collapse
Affiliation(s)
- Elizabeth Vincan
- Cancer Biology Laboratory, Department of Anatomy and Cell Biology, University of Melbourne, Australia.
| | | | | | | | | |
Collapse
|
23
|
Abstract
Cells in tissues do not exist as isolated entities but are part of the three-dimensional tissue architecture. Consequently, some aspects of cell behaviour cannot be mimicked by simple in vitro monolayer culture systems. Moreover, cell shape and behaviour is not rigid but is dynamic and can be regulated by intrinsic and extrinsic factors. For example, tumour cells in epithelium-derived cancer such as colorectal cancer often retain significant features of the colonic mucosa. However, as the tumour progresses, the morphology of the tumour cells often undergoes a transition from an epithelial morphology to a mesenchymal morphology. This transition is important as it signifies a change in the tumour phenotype to a more aggressive, invasive, and eventually metastatic phenotype. In vitro models that allow the study of this transition are needed. One such model is the LIM1863 colon carcinoma cells that normally grow as organoids but can be adapted to efficiently undergo an epithelial to mesenchymal transition that can be reversed. This system has allowed the study of the genes such as Frizzled 7 that are involved in this dynamic and reversible epithelial to mesenchymal transition.
Collapse
|
24
|
Epithelial to mesenchymal transition of a primary prostate cell line with switches of cell adhesion modules but without malignant transformation. PLoS One 2008; 3:e3368. [PMID: 18852876 PMCID: PMC2557125 DOI: 10.1371/journal.pone.0003368] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Accepted: 09/03/2008] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Epithelial to mesenchymal transition (EMT) has been connected with cancer progression in vivo and the generation of more aggressive cancer cell lines in vitro. EMT has been induced in prostate cancer cell lines, but has previously not been shown in primary prostate cells. The role of EMT in malignant transformation has not been clarified. METHODOLOGY/PRINCIPAL FINDINGS In a transformation experiment when selecting for cells with loss of contact inhibition, the immortalized prostate primary epithelial cell line, EP156T, was observed to undergo EMT accompanied by loss of contact inhibition after about 12 weeks in continuous culture. The changed new cells were named EPT1. EMT of EPT1 was characterized by striking morphological changes and increased invasion and migration compared with the original EP156T cells. Gene expression profiling showed extensively decreased epithelial markers and increased mesenchymal markers in EPT1 cells, as well as pronounced switches of gene expression modules involved in cell adhesion and attachment. Transformation assays showed that EPT1 cells were sensitive to serum or growth factor withdrawal. Most importantly, EPT1 cells were not able to grow in an anchorage-independent way in soft agar, which is considered a critical feature of malignant transformation. CONCLUSIONS/SIGNIFICANCE This work for the first time established an EMT model from primary prostate cells. The results show that EMT can be activated as a coordinated gene expression program in association with early steps of transformation. The model allows a clearer identification of the molecular mechanisms of EMT and its potential role in malignant transformation.
Collapse
|
25
|
Kim JH, Kim MA, Lee HS, Kim WH. Comparative analysis of protein expressions in primary and metastatic gastric carcinomas. Hum Pathol 2008; 40:314-22. [PMID: 18835621 DOI: 10.1016/j.humpath.2008.07.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 07/07/2008] [Accepted: 07/22/2008] [Indexed: 11/28/2022]
Abstract
Because metastatic cancers are derived from their primary counterparts, their molecular profiles could reasonably be expected to be similar to those of primary cancers. However, this expectation has been proven to be untrue in several human cancers. To explore protein expressional differences in primary and metastatic gastric carcinoma, we evaluated the expressions of 32 tumor-associated proteins in 250 pairs of primary and metastatic gastric carcinoma tissues by immunohistochemistry using tissue array slides. In metastatic gastric carcinomas, the expressions of epidermal growth factor receptor, c-erbB2, and trefoil factor 1(TFF-1) were higher and those of beta-catenin, E-cadherin, fragile histone triad gene (FHIT), glutathione S transferase-pi (GST-pi), kangai 1 (KAI1), and nuclear factor-kappaB (NF-kappaB) were lower than in primary gastric carcinomas. Furthermore, the expressions of beta-catenin, E-cadherin, KAI1, and NF-kappaB were associated with an advanced T and combined stage. In addition, the loss of E-cadherin expression during lymph node metastasis or E-cadherin immunonegativity in metastatic lesions and epidermal growth factor receptor expression in primary gastric carcinomas were independently associated with a poor prognosis by multivariate analysis. In conclusion, the expression of some tumor-associated proteins and their prognostic significance in metastatic gastric carcinomas differ from those in primary tumors. Consequently, analysis of both metastatic gastric carcinomas and their primary counterparts may be required to fully determine the molecular characteristics of node-positive gastric carcinoma.
Collapse
Affiliation(s)
- Ji Hun Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | | | | | | |
Collapse
|
26
|
Zhau HE, Odero-Marah V, Lue HW, Nomura T, Wang R, Chu G, Liu ZR, Zhou BP, Huang WC, Chung LWK. Epithelial to mesenchymal transition (EMT) in human prostate cancer: lessons learned from ARCaP model. Clin Exp Metastasis 2008; 25:601-10. [PMID: 18535913 PMCID: PMC3050737 DOI: 10.1007/s10585-008-9183-1] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Accepted: 05/14/2008] [Indexed: 12/24/2022]
Abstract
Androgen refractory cancer of the prostate (ARCaP) cells contain androgen receptor (AR) and synthesize and secrete prostate specific antigen (PSA). We isolated epithelia-like ARCaP(E) from parental ARCaP cells and induced them to undergo epithelial-mesenchymal transition (EMT) by exposing these cells to soluble factors including TGFbeta1 plus EGF, IGF-1, beta2-microglobulin (beta2-m), or a bone microenvironment. The molecular and behavioral characteristics of the resultant ARCaP(M) were characterized extensively in comparison to the parental ARCaP(E) cells. In addition to expressing mesenchymal biomarkers, ARCaP(M) gained 100% incidence of bone metastasis. ARCaP(M) cells express receptor activator of NF-kappaB ligand (RANKL), which was shown to increase tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts in culture, and when metastatic to bone in vivo. We provide evidence that RANKL expression was promoted by increased cell signaling mediated by the activation of Stat3-Snail-LIV-1. RANKL expressed by ARCaP(M) cells is functional both in vitro and in vivo. The lesson we learned from the ARCaP model of EMT is that activation of a specific cell signaling pathway by soluble factors can lead to increased bone turnover, mediated by enhanced RANKL expression by tumor cells, which is implicated in the high incidence of prostate cancer bone colonization. The ARCaP EMT model is highly attractive for developing new therapeutic agents to treat prostate cancer bone metastasis.
Collapse
Affiliation(s)
- Haiyen E. Zhau
- Molecular Urology & Therapeutics Program, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, Suite 5107, Atlanta, GA 30322, USA
| | - Valerie Odero-Marah
- Molecular Urology & Therapeutics Program, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, Suite 5107, Atlanta, GA 30322, USA
| | - Hui-Wen Lue
- Molecular Urology & Therapeutics Program, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, Suite 5107, Atlanta, GA 30322, USA. Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Takeo Nomura
- Molecular Urology & Therapeutics Program, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, Suite 5107, Atlanta, GA 30322, USA
| | - Ruoxiang Wang
- Molecular Urology & Therapeutics Program, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, Suite 5107, Atlanta, GA 30322, USA
| | - Gina Chu
- Molecular Urology & Therapeutics Program, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, Suite 5107, Atlanta, GA 30322, USA. Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Zhi-Ren Liu
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Binhua P. Zhou
- Sealy Center for Cancer Cell Biology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Wen-Chin Huang
- Molecular Urology & Therapeutics Program, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, Suite 5107, Atlanta, GA 30322, USA
| | - Leland W. K. Chung
- Molecular Urology & Therapeutics Program, Department of Urology, Emory University School of Medicine, 1365B Clifton Road, Suite 5107, Atlanta, GA 30322, USA
| |
Collapse
|