51
|
Yoshida GJ, Saya H. EpCAM expression in the prostate cancer makes the difference in the response to growth factors. Biochem Biophys Res Commun 2013; 443:239-45. [PMID: 24309103 DOI: 10.1016/j.bbrc.2013.11.093] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 11/23/2013] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Epithelial cell adhesion molecule (EpCAM) is expressed in tumors with an epithelial cell of origin, in a heterogeneous manner. Prostate cancer stem-like cells highly express EpCAM. However, little is known about how EpCAM is involved in the ability of cells to adapt to micro-environmental changes in available growth factors, which is one of the essential biological phenotypes of cancer stem-like cells (CSCs). METHODS EpCAM-high and EpCAM-low subpopulations of cells were established from the prostate cancer cell line PC-3. Signal transductions in response to serum starvation, and on the exposure to EGF ligand or the specific inhibitor were analyzed in terms. Furthermore, we analyzed the expression level of amino acid transporters which contribute to the activation of mTOR signal between the two subgroups. RESULTS EpCAM-high and EpCAM-low PC-3 subpopulations showed markedly different responses to serum starvation. EpCAM expression was positively correlated with activation of the mTOR and epithelial growth factor receptor (EGFR) signaling pathways. Furthermore, AMP-activated protein kinase (AMPK) was gradually de-activated in EpCAM-low PC-3 cells in the absence of serum. CONCLUSIONS EpCAM regulates the AMPK signaling pathway, essential for the response to growth factors characterized by EGF. LAT1, the amino acid transporter stabilized at the cellular membrane by EpCAM, is likely to be responsible for the difference in the susceptibility to EGF between EpCAM-high and EpCAM-low PC-3 cells.
Collapse
Affiliation(s)
- Go J Yoshida
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
52
|
Maghzal N, Kayali HA, Rohani N, Kajava AV, Fagotto F. EpCAM controls actomyosin contractility and cell adhesion by direct inhibition of PKC. Dev Cell 2013; 27:263-77. [PMID: 24183651 DOI: 10.1016/j.devcel.2013.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 07/03/2013] [Accepted: 10/07/2013] [Indexed: 01/06/2023]
Abstract
Epithelial cell adhesion molecule (EpCAM) is a cell-surface protein highly expressed in embryonic tissues and in malignant carcinomas. We report that EpCAM acts as a potent inhibitor of novel protein kinase C (nPKC) in both embryos and cancer cells. We observed dramatic effects of loss of EpCAM on amphibian embryonic tissues, which include sequentially strong overstimulation of PKC activity and of the Erk pathway, leading to exacerbated myosin contractility, loss of cadherin-mediated adhesion, tissue dissociation, and, ultimately, cell death. We show that PKC inhibition is caused by a short segment of the EpCAM cytoplasmic tail. This motif resembles the pseudosubstrate inhibitory domains of PKCs and binds nPKCs with high affinity. A bioinformatics search reveals the existence of similar motifs in other plasma membrane proteins, most of which are cell-cell adhesion molecules. Thus, direct inhibition of PKC by EpCAM represents a general mode of regulation of signal transduction by cell-surface proteins.
Collapse
Affiliation(s)
- Nadim Maghzal
- Department of Biology, McGill University, Montreal, H3A1B1 Quebec, Canada
| | | | | | | | | |
Collapse
|
53
|
Ni J, Cozzi P, Hao J, Beretov J, Chang L, Duan W, Shigdar S, Delprado W, Graham P, Bucci J, Kearsley J, Li Y. Epithelial cell adhesion molecule (EpCAM) is associated with prostate cancer metastasis and chemo/radioresistance via the PI3K/Akt/mTOR signaling pathway. Int J Biochem Cell Biol 2013; 45:2736-48. [PMID: 24076216 DOI: 10.1016/j.biocel.2013.09.008] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/06/2013] [Accepted: 09/14/2013] [Indexed: 12/21/2022]
Abstract
Prostate cancer (CaP) is the second leading malignancy in men. The role of epithelial cell adhesion molecule (EpCAM), also known as CD326, in CaP progression and therapeutic resistance is still uncertain. Here, we aimed to investigate the roles of EpCAM in CaP metastasis and chemo/radioresistance. Expression of EpCAM in CaP cell lines and human CaP tissues was assessed using immunofluorescence and immunohistochemistry, respectively. EpCAM was knocked down (KD) in PC-3, DU145 and LNCaP-C4-2B cells using small interfering RNA (siRNA), and KD results were confirmed by confocal microscope, Western blotting and quantitative real time polymerase chain reaction (qRT-PCR). Cell growth was evaluated by proliferation and colony formation assays. The invasive potential was assessed using a matrigel chamber assay. Tumorigenesis potential was measured by a sphere formation assay. Chemo-/radiosensitivity were measured using a colony formation assay. Over-expression of EpCAM was found in primary CaP tissues and lymph node metastases including cancer cells and surrounding stromal cells. KD of EpCAM suppressed CaP proliferation and invasive ability, reduced sphere formation, enhanced chemo-/radiosensitivity, and down-regulated E-cadherin, p-Akt, p-mTOR, p-4EBP1 and p-S6K expression in CaP cells. Our findings suggest that EpCAM plays an important role in CaP proliferation, invasion, metastasis and chemo-/radioresistance associated with the activation of the PI3K/Akt/mTOR signaling pathway and is a novel therapeutic target to sensitize CaP cells to chemo-/radiotherapy.
Collapse
Affiliation(s)
- Jie Ni
- Cancer Care Centre and Prostate Cancer Institute, St George Hospital, Kogarah, NSW 2217, Australia; St George Clinical School, University of New South Wales (UNSW), Kensington, NSW 2052, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Abstract
Despite significant advances in surgery, radiotherapy and chemotherapy to treat prostate cancer (CaP), many patients die of secondary disease (metastases). Current therapeutic approaches are limited, and there is no cure for metastatic castration-resistant prostate cancer (CRPC). Epithelial cell adhesion molecule (EpCAM, also known as CD326) is a transmembrane glycoprotein that is highly expressed in rapidly proliferating carcinomas and plays an important role in the prevention of cell-cell adhesion, cell signalling, migration, proliferation and differentiation. Stably and highly expressed EpCAM has been found in primary CaP tissues, effusions and CaP metastases, making it an ideal candidate of tumour-associated antigen to detect metastasis of CaP cells in the circulation as well as a promising therapeutic target to control metastatic CRPC disease. In this review, we discuss the implications of the newly identified roles of EpCAM in terms of its diagnostic and metastatic relevance to CaP. We also summarize EpCAM expression in human CaP and EpCAM-mediated signalling pathways in cancer metastasis. Finally, emerging and innovative approaches to the management of the disease and expanding potential therapeutic applications of EpCAM for targeted strategies in future CaP therapy will be explored.
Collapse
|
55
|
Hao H, Zhen Y, Wang Z, Chen F, Xie X. A novel therapeutic drug for colon cancer: EpCAM scFv-truncated protamine (tp)-siRNA. Cell Biol Int 2013; 37:860-4. [PMID: 23576466 DOI: 10.1002/cbin.10112] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 03/29/2012] [Indexed: 12/15/2022]
Abstract
Colon cancer is a type of malignant tumor that causes considerable mortality worldwide. Epithelial cellular adhesion molecule (EpCAM), a tumor-associated antigen of colon tumors, is a target for colon cancer therapy. EpCAM-specific monoclonal antibodies (mAbs) have been applied in human colon cancer since the 1990s; however, the therapeutic effects are limited. EpCAM activates nuclear signaling pathways by releasing its intracellular domain (EpICD). The released EpICD stimulates the Wnt/β-catenin signaling pathway, which is also strongly associated with tumorigenesis. EpCAM is also a target gene of the Wnt/β-catenin signaling pathway. EpCAM and the Wnt/β-catenin signaling pathway form a functional interaction cycle in colon cancer. Thus, we propose a new therapeutic drug for colon cancer: an EpCAM single-chain fragment variable antibody (scFv)-truncated protamine-siRNA. EpCAM scFv can recognize and bind colon cancer cells through its EpCAM antigen activity. Furthermore, the specific siRNA transferred into colon cancer cells specifically inhibits Wnt/β-catenin signal transmission. Therefore, this new drug may efficiently interrupt the functional cycle between EpCAM and Wnt/β-catenin signaling and be an effective therapeutic strategy for colon cancer.
Collapse
Affiliation(s)
- Huiwen Hao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, 710069, China
| | | | | | | | | |
Collapse
|
56
|
Bidirectional modulation of endogenous EpCAM expression to unravel its function in ovarian cancer. Br J Cancer 2013; 108:881-6. [PMID: 23403823 PMCID: PMC3590680 DOI: 10.1038/bjc.2013.45] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background: The epithelial cell adhesion molecule (EpCAM) is overexpressed on most carcinomas. Dependent on the tumour type, its overexpression is either associated with improved or worse patient survival. For ovarian cancer, however, the role of EpCAM remains unclear. Methods: Cell survival of ovarian cancer cell lines was studied after induction or repression of endogenous EpCAM expression using siRNA/cDNA or artificial transcription factors (ATF) consisting of engineered zinc-fingers fused to either a transcriptional activator or repressor domain. Results: Two ATFs were selected as the most potent down- and upregulator, showing at least a two-fold alteration of EpCAM protein expression compared with control. Downregulation of EpCAM expression resulted in growth inhibition in breast cancer, but showed no effect on cell growth in ovarian cancer. Induction or further upregulation of EpCAM expression decreased ovarian cancer cell survival. Conclusion: The bidirectional ATF-based approach is uniquely suited to study cell-type-specific biological effects of EpCAM expression. Using this approach, the oncogenic function of EpCAM in breast cancer was confirmed. Despite its value as a diagnostic marker and for immunotherapy, EpCAM does not seem to represent a therapeutic target for gene expression silencing in ovarian cancer.
Collapse
|
57
|
Santana SM, Liu H, Bander NH, Gleghorn JP, Kirby BJ. Immunocapture of prostate cancer cells by use of anti-PSMA antibodies in microdevices. Biomed Microdevices 2012; 14:401-7. [PMID: 22143878 DOI: 10.1007/s10544-011-9616-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patients suffering from cancer can shed tumor cells into the bloodstream, leading to one of the most important mechanisms of metastasis. As such, the capture of these cells is of great interest. Circulating tumor cells are typically extracted from circulation through positive selection with the epithelial cell-adhesion molecule (EpCAM), leading to currently unknown biases when cells are undergoing epithelial-to-mesenchymal transition. For prostate cancer, prostate-specific membrane antigen (PSMA) presents a compelling target for immunocapture, as PSMA levels increase in higher-grade cancers and metastatic disease and are specific to the prostate epithelium. This study uses monoclonal antibodies J591 and J415-antibodies that are highly specific for intact extracellular domains of PSMA on live cells-in microfluidic devices for the capture of LNCaPs, a PSMA-expressing immortalized prostate cancer cell line, over a range of concentrations and shear stresses relevant to immunocapture. Our results show that J591 outperforms J415 and a mix of the two for prostate cancer capture, and that capture performance saturates following incubation with antibody concentrations of 10 micrograms per milliliter.
Collapse
Affiliation(s)
- Steven M Santana
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, 245 Upson Hall, Ithaca, NY 14853, USA.
| | | | | | | | | |
Collapse
|
58
|
Kirby BJ, Jodari M, Loftus MS, Gakhar G, Pratt ED, Chanel-Vos C, Gleghorn JP, Santana SM, Liu H, Smith JP, Navarro VN, Tagawa ST, Bander NH, Nanus DM, Giannakakou P. Functional characterization of circulating tumor cells with a prostate-cancer-specific microfluidic device. PLoS One 2012; 7:e35976. [PMID: 22558290 PMCID: PMC3338784 DOI: 10.1371/journal.pone.0035976] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 03/23/2012] [Indexed: 01/21/2023] Open
Abstract
Cancer metastasis accounts for the majority of cancer-related deaths owing to poor response to anticancer therapies. Molecular understanding of metastasis-associated drug resistance remains elusive due to the scarcity of available tumor tissue. Isolation of circulating tumor cells (CTCs) from the peripheral blood of patients has emerged as a valid alternative source of tumor tissue that can be subjected to molecular characterization. However, issues with low purity and sensitivity have impeded adoption to clinical practice. Here we report a novel method to capture and molecularly characterize CTCs isolated from castrate-resistant prostate cancer patients (CRPC) receiving taxane chemotherapy. We have developed a geometrically enhanced differential immunocapture (GEDI) microfluidic device that combines an anti-prostate specific membrane antigen (PSMA) antibody with a 3D geometry that captures CTCs while minimizing nonspecific leukocyte adhesion. Enumeration of GEDI-captured CTCs (defined as intact, nucleated PSMA+/CD45- cells) revealed a median of 54 cells per ml identified in CRPC patients versus 3 in healthy donors. Direct comparison with the commercially available CellSearch® revealed a 2-400 fold higher sensitivity achieved with the GEDI device. Confocal microscopy of patient-derived GEDI-captured CTCs identified the TMPRSS2:ERG fusion protein, while sequencing identified specific androgen receptor point mutation (T868A) in blood samples spiked with only 50 PC C4-2 cells. On-chip treatment of patient-derived CTCs with docetaxel and paclitaxel allowed monitoring of drug-target engagement by means of microtubule bundling. CTCs isolated from docetaxel-resistant CRPC patients did not show any evidence of drug activity. These measurements constitute the first functional assays of drug-target engagement in living circulating tumor cells and therefore have the potential to enable longitudinal monitoring of target response and inform the development of new anticancer agents.
Collapse
Affiliation(s)
- Brian J. Kirby
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, United States of America
- Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Mona Jodari
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - Matthew S. Loftus
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - Gunjan Gakhar
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - Erica D. Pratt
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, United States of America
- Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Chantal Chanel-Vos
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - Jason P. Gleghorn
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, United States of America
- Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Steven M. Santana
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, United States of America
- Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - He Liu
- Department of Urology, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - James P. Smith
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, United States of America
- Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Vicente N. Navarro
- Department of Urology, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - Scott T. Tagawa
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - Neil H. Bander
- Department of Urology, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - David M. Nanus
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
- Department of Urology, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| | - Paraskevi Giannakakou
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York, United States of America
| |
Collapse
|
59
|
Sankpal NV, Mayfield JD, Willman MW, Fleming TP, Gillanders WE. Activator protein 1 (AP-1) contributes to EpCAM-dependent breast cancer invasion. Breast Cancer Res 2011; 13:R124. [PMID: 22132731 PMCID: PMC3326566 DOI: 10.1186/bcr3070] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 11/02/2011] [Accepted: 12/01/2011] [Indexed: 01/24/2023] Open
Abstract
Introduction EpCAM is a cell-surface glycoprotein that is overexpressed in the majority of epithelial carcinomas. However, the functional role of EpCAM in regulating cancer invasion remains controversial, and the mechanism(s) underlying EpCAM-mediated regulation of breast cancer invasion remain to be defined. Methods EpCAM expression was manipulated in breast cancer cell lines using RNA interference and cDNA expression constructs. Recombinant EpCAM was used to rescue EpCAM signaling following specific ablation of EpCAM. Protein and gene expression, invasion, transcription factor activity, and protein phosphorylation were measured using standard molecular biology techniques. Results In loss-of-function, and gain-of-function experiments we demonstrate that EpCAM expression is associated with increased breast cancer invasion in vitro and in vivo. We demonstrate further that specific ablation of EpCAM expression is associated with decreased activator protein-1 (AP-1) transcription factor activity. Phosphoprotein analyses confirm that specific ablation of EpCAM is associated with decreased phosphorylation of the AP-1 subunit c-Jun. Recombinant soluble extracellular EpCAM (rEpCAM) is able to rescue invasion, AP-1 transcription factor activity, and c-Jun phosphorylation in a dose-dependent fashion. Pharmacologic inhibitors, and constitutively active constructs of the c-Jun N-terminal kinase (JNK) signal transduction pathway, suggest that the impact of EpCAM expression on AP-1 transcription factor activity is mediated through the JNK pathway. In functional rescue experiments, forced expression of c-Jun rescues invasion in breast cancer cells following specific ablation of EpCAM. Conclusions These data demonstrate for the first time that EpCAM expression can influence the JNK/AP-1 signal transduction pathway, and suggest that modulation of AP-1 transcription factor activity contributes to EpCAM-dependent breast cancer invasion. These data have important implications for the design and application of molecular therapies targeting EpCAM.
Collapse
Affiliation(s)
- Narendra V Sankpal
- Department of Surgery, Washington University School of Medicine, Campus Box 8109, 660 South Euclid Avenue, St, Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|