1
|
Monti N, Antinori D, Proietti S, Piombarolo A, Querqui A, Lentini G, Liguoro D, Aventaggiato M, Lucarelli M, Pensotti A, Giuliani A, Tafani M, Fuso A, Bizzarri M. miRNAs from Zebrafish Embryo Extracts Inhibit Breast Cancer Invasiveness and Migration by Modulating miR-218-5p/PI3K Pathway. Int J Mol Sci 2025; 26:3812. [PMID: 40332412 PMCID: PMC12027887 DOI: 10.3390/ijms26083812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/05/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Herein, we demonstrate that soluble factors extracted from the distinct phases of the development of zebrafish embryos (ZFEs) exhibit a specific miRNA profile. We removed proteins and concentrated miRNAs in different phase-related samples, which we investigated further. We observed that ZFEs modulate miRNA expression in both normal and cancerous breast cells, significantly inhibiting the invasiveness and motility of triple-negative breast cancer cells. Namely, ZFEs reactivate the synthesis of miR-218-5p in cancerous cells, leading to the downregulation of PI3K, which consequently alters the distribution of phosphoinositides (such as PIP2/PIP3). Moreover, the silencing of miR-218-5p abolished the ZFE effects. Restoring a proper PIP2/PIP3 ratio is crucial for promoting the regression of the malignant phenotype. Phenotypic reversion follows the extensive cytoskeleton rearrangement and the re-emergence of E-cadherin/β-catenin complexes. In addition, ZFEs antagonize the Epithelial Mesenchymal Transition (EMT) by modulating several pathways, including the TCTP-p53 axis. Overall, these results show that embryo extracts enriched with fish miRNAs reactivate endogenous miR-218-5p in cancerous cells, which in turn downregulates critical pathways involved in tumor progression and metastasis.
Collapse
Affiliation(s)
- Noemi Monti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
- System Biology Group Lab, Sapienza University of Rome, 00161 Rome, Italy;
| | - Daniele Antinori
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
| | - Sara Proietti
- System Biology Group Lab, Sapienza University of Rome, 00161 Rome, Italy;
| | - Aurora Piombarolo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
- System Biology Group Lab, Sapienza University of Rome, 00161 Rome, Italy;
| | - Alessandro Querqui
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
- System Biology Group Lab, Sapienza University of Rome, 00161 Rome, Italy;
| | - Guglielmo Lentini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
- System Biology Group Lab, Sapienza University of Rome, 00161 Rome, Italy;
| | - Domenico Liguoro
- SAFU Laboratory, Department of Research, Advanced Diagnostics and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Michele Aventaggiato
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy
| | - Andrea Pensotti
- Research Unit of Philosophy of Science and Human Development, University Campus Bio-Medico of Rome, 00128 Rome, Italy;
| | - Alessandro Giuliani
- Environment and Health Department, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
- CRiN, Center for Research in Neurobiology D. Bovet, Sapienza University of Rome, 00161 Rome, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (D.A.); (A.P.); (A.Q.); (G.L.); (M.A.); (M.L.); (M.T.); (A.F.)
- System Biology Group Lab, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
2
|
Files R, Cardoso C, Prada J, Silva F, Pires I. Syndecan-1 and E-Cadherin Expression in Canine Cutaneous Squamous Cell Carcinoma. Vet Sci 2024; 11:652. [PMID: 39728992 DOI: 10.3390/vetsci11120652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) in dogs is a locally invasive tumor that typically occurs in areas of poorly pigmented skin due to sun exposure. Identifying new biomarkers, such as syndecan-1 (CD138) and E-cadherin, is fundamental for tumor diagnosis and prognosis. Dysregulation of syndecan-1, expressed in epithelial tissue, fibroblasts, and plasma cells, is associated with poor prognosis in several types of cancer. Similarly, E-cadherin, which plays a crucial role in cell adhesion and epithelial functionality, is also linked to adverse outcomes. This study evaluated the expression of syndecan-1 and E-cadherin in 47 cases of canine cutaneous squamous cell carcinoma. The results showed that the intensity of syndecan-1 decreased with increasing tumor aggressiveness, and its presence in the stroma was significantly associated with tumor grade. E-cadherin also demonstrated a decrease in intensity with increasing malignancy. However, the association between syndecan-1 and E-cadherin was not statistically significant. E-cadherin reduction and stromal syndecan-1 positivity seem to be associated with tumor aggressiveness in canine cutaneous squamous cell carcinoma. Further studies are needed to explore their roles in tumor progression.
Collapse
Affiliation(s)
- Rita Files
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Cláudia Cardoso
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Justina Prada
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Filipe Silva
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Isabel Pires
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| |
Collapse
|
3
|
Antonova L, Paramanthan P, Falls T, Wedge ME, Mayer J, Sekhon HS, McPherson J, Denroche RE, Gallinger S, Bell JC, Ilkow CS, Chatterjee A. Molecular Characterization and Xenotransplantation of Pancreatic Cancer Using Endoscopic Ultrasound-Guided Fine Needle Aspiration (EUS-FNA). Cancers (Basel) 2024; 16:2721. [PMID: 39123450 PMCID: PMC11311391 DOI: 10.3390/cancers16152721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Pancreatic cancer has one of the worst prognoses among all malignancies and few available treatment options. Patient-derived xenografts can be used to develop personalized therapy for pancreatic cancer. Endoscopic ultrasound fine-needle aspiration (EUS-FNA) may provide a powerful alternative to surgery for obtaining sufficient tissue for the establishment of patient-derived xenografts. In this study, EUS-FNA samples were obtained for 30 patients referred to the Ottawa Hospital, Ottawa, Ontario, Canada. These samples were used for xenotransplantation in NOD-SCID mice and for genetic analyses. The gene expression of pancreatic-cancer-relevant genes in xenograft tumors was examined by immunohistochemistry. Targeted sequencing of both the patient-derived tumors and xenograft tumors was performed. The xenografts' susceptibility to oncolytic virus infection was studied by infecting xenograft-derived cells with VSV∆51-GFP. The xenograft take rate was found to be 75.9% for passage 1 and 100% for passage 2. Eighty percent of patient tumor samples were successfully sequenced to a high depth for 42 cancer genes. Xenograft histological characteristics and marker expression were maintained between passages. All tested xenograft samples were susceptible to oncoviral infection. We found that EUS-FNA is an accessible, minimally invasive technique that can be used to acquire adequate pancreatic cancer tissue for the generation of patient-derived xenografts and for genetic sequencing.
Collapse
Affiliation(s)
- Lilia Antonova
- Department of Otolaryngology-Head and Neck Surgery, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Piriya Paramanthan
- Division of Gastroenterology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Theresa Falls
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Marie-Eve Wedge
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Justin Mayer
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Harman S. Sekhon
- Division of Anatomic Pathology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada;
| | - John McPherson
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | | | - Steven Gallinger
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - John Cameron Bell
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Carolina S. Ilkow
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Avijit Chatterjee
- Division of Gastroenterology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
4
|
Shi L, Zhang D, Han H, Zhang L, Li S, Yang F, He C. HOTAIR knockdown impairs metastasis of cervical cancer cells by down-regulating metastasis-related genes. J OBSTET GYNAECOL 2023; 43:2181060. [PMID: 36972141 DOI: 10.1080/01443615.2023.2181060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
This study investigated the role of LncRNA HOTAIR knockdown in the biological impacts on cervical cancer cells. The HOTAIR gene in two human cervical cancer cell lines was silenced with small interfering (si) RNA siHOTAIR. Proliferation, apoptosis, migration and invasion of cells were assessed following the knockdown. The expressions of Notch1, EpCAM, E-cadherin, vimentin and STAT3 were assessed using qRT-PCR and Western blotting analysis. Compared with controls, HOTAIR levels were reduced significantly, the OD values of cells were significantly decreased in proliferation assays, cell apoptosis was significantly increased, cell migration and invasion were significantly reduced after HOTAIR knockdown. Molecular analysis showed that Notch1, EpCAM, vimentin and STAT3 expressions were decreased significantly, while the expression of E-cadherin was significantly increased after HOTAIR knockdown. Rescue experiments further confirmed that Notch1 and STAT3 were involved in siHOTAIR-mediated reduction of migration and invasion of cervical cancer cells.IMPACT STATEMENTWhat is already known on this subject? Long non-coding RNAs including HOTAIR, is implicated in occurrence and development of cancer and have been explored to develop new therapeutic options for cancer.What do the results of this study add? HOTAIR silencing significantly reduces the viability and migration ability of cells and induces cell apoptosis, adding experimental data supporting the potential use of HOTAIR specific-siRNA as a therapeutic avenue for the cancer.What are the implications of these findings for clinical practice and/or further research? The finding from this study would help develop clinically applicable therapeutic avenues for the cancer and identify new treatment targets in the relevant pathways leading to new drugs or treatments.
Collapse
Affiliation(s)
- Lei Shi
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Dehui Zhang
- Department of Oncology, Daqing Oilfield General Hospital and Huiren Cancer Hospital, Daqing, P.R. China
| | - Huijuan Han
- Department of Obstetrics and Gynecology, the 962 Hospital, Joint Logistics Support of the Chinese People's Liberation Army, Harbin, P.R. China
| | - Liangyu Zhang
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Sirui Li
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Fang Yang
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Caijun He
- Department of Obstetrics and Gynecology, the 962 Hospital, Joint Logistics Support of the Chinese People's Liberation Army, Harbin, P.R. China
| |
Collapse
|
5
|
Chakravarthi S, Karikalan B. Molecular Biomarkers for Lung Adenocarcinoma: A Short Review. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716666200724164654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer is a disease with higher death rates and is responsible for around 2 million
deaths per year worldwide. Recently, several breakthroughs have been made in the field of lung
cancer that has led to a revolution in the management of lung cancer patients. Identification of
molecular markers and the implication of respective targeted therapies has been a great success in
the treatment of lung adenocarcinoma patients. Despite the fact that targeted therapy of lung adenocarcinomas
represents one of the significant milestones in the treatment of lung cancer that resulted
in increased survival rates even in advanced stages, the mortality rates of lung cancer still remain
to be significantly high. This warrants further research for gaining better insights into molecular alterations
that can lead to newer innovations in targeted drug therapy towards lung adenocarcinoma.
In this review, we briefly summarized the literature on molecular markers that are already in use.
We also consolidated newer molecular markers that are under study with the potential for being targeted
for therapies in future.
Collapse
|
6
|
AlMusawi S, Ahmed M, Nateri AS. Understanding cell-cell communication and signaling in the colorectal cancer microenvironment. Clin Transl Med 2021; 11:e308. [PMID: 33635003 PMCID: PMC7868082 DOI: 10.1002/ctm2.308] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/31/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Carcinomas are complex heterocellular systems containing epithelial cancer cells, stromal fibroblasts, and multiple immune cell-types. Cell-cell communication between these tumor microenvironments (TME) and cells drives cancer progression and influences response to existing therapies. In order to provide better treatments for patients, we must understand how various cell-types collaborate within the TME to drive cancer and consider the multiple signals present between and within different cancer types. To investigate how tissues function, we need a model to measure both how signals are transferred between cells and how that information is processed within cells. The interplay of collaboration between different cell-types requires cell-cell communication. This article aims to review the current in vitro and in vivo mono-cellular and multi-cellular cultures models of colorectal cancer (CRC), and to explore how they can be used for single-cell multi-omics approaches for isolating multiple types of molecules from a single-cell required for cell-cell communication to distinguish cancer cells from normal cells. Integrating the existing single-cell signaling measurements and models, and through understanding the cell identity and how different cell types communicate, will help predict drug sensitivities in tumor cells and between- and within-patients responses.
Collapse
Affiliation(s)
- Shaikha AlMusawi
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Division of Cancer & Stem Cells, School of MedicineUniversity of NottinghamNottinghamUK
| | - Mehreen Ahmed
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Division of Cancer & Stem Cells, School of MedicineUniversity of NottinghamNottinghamUK
- Department of Laboratory Medicine, Division of Translational Cancer ResearchLund UniversityLundSweden
| | - Abdolrahman S. Nateri
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Division of Cancer & Stem Cells, School of MedicineUniversity of NottinghamNottinghamUK
| |
Collapse
|
7
|
Tiwari R, Manzar N, Ateeq B. Dynamics of Cellular Plasticity in Prostate Cancer Progression. Front Mol Biosci 2020; 7:130. [PMID: 32754615 PMCID: PMC7365877 DOI: 10.3389/fmolb.2020.00130] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
Despite the current advances in the treatment for prostate cancer, the patients often develop resistance to the conventional therapeutic interventions. Therapy-induced drug resistance and tumor progression have been associated with cellular plasticity acquired due to reprogramming at the molecular and phenotypic levels. The plasticity of the tumor cells is mainly governed by two factors: cell-intrinsic and cell-extrinsic. The cell-intrinsic factors involve alteration in the genetic or epigenetic regulators, while cell-extrinsic factors include microenvironmental cues and drug-induced selective pressure. Epithelial-mesenchymal transition (EMT) and stemness are two important hallmarks that dictate cellular plasticity in multiple cancer types including prostate. Emerging evidence has also pinpointed the role of tumor cell plasticity in driving anti-androgen induced neuroendocrine prostate cancer (NEPC), a lethal and therapy-resistant subtype. In this review, we discuss the role of cellular plasticity manifested due to genetic, epigenetic alterations and cues from the tumor microenvironment, and their role in driving therapy resistant prostate cancer.
Collapse
Affiliation(s)
| | | | - Bushra Ateeq
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
8
|
Ye T, Li J, Sun Z, Liu D, Zeng B, Zhao Q, Wang J, Xing HR. Cdh1 functions as an oncogene by inducing self-renewal of lung cancer stem-like cells via oncogenic pathways. Int J Biol Sci 2020; 16:447-459. [PMID: 32015681 PMCID: PMC6990901 DOI: 10.7150/ijbs.38672] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/04/2019] [Indexed: 01/01/2023] Open
Abstract
The mortality rate of lung cancer remains the highest amongst all cancers despite of new therapeutic developments. While cancer stem cells (CSCs) may play a pivotal role in cancer, mechanisms underlying CSCs self-renewal and their relevance to cancer progression have not been clearly elucidated due to the lack of reliable and stable CSC cellular models. In the present study, we unveiled the novel oncogene function of cadherin 1 (Cdh1) via bioinformatic analysis in a broad spectrum of human cancers including lung adenocarcinoma (LUAD), adding a new dimension to the widely reported tumor suppressor function of Cdh1. Experimentally, we show for the first time that Cdh1 promotes the self-renewal of lung CSCs, consistent with its function in embryonic and normal stem cells. Using the LLC-Symmetric Division (LLC-SD) model, we have revealed an intricate cross-talk between the oncogenic pathway and stem cell pathway in which Cdh1 functions as an oncogene by promoting lung CSC renewal via the activation of the Phosphoinositide 3-kinase (PI3K) and inhibition of Mitogen-activated protein kinase (MAPK) pathways, respectively. In summary, this study has provided evidence demonstrating effective utilization of the normal stem cell renewal mechanisms by CSCs to promote oncogenesis and progression.
Collapse
Affiliation(s)
- Ting Ye
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China.,Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingyuan Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Zhiwei Sun
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Doudou Liu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Bin Zeng
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Qiting Zhao
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Jianyu Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - H Rosie Xing
- Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China.,College of Biomedical Engineering, State Key Laboratory of Ultrasound Engineering in Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Laganà AS, Garzon S, Götte M, Viganò P, Franchi M, Ghezzi F, Martin DC. The Pathogenesis of Endometriosis: Molecular and Cell Biology Insights. Int J Mol Sci 2019; 20:E5615. [PMID: 31717614 PMCID: PMC6888544 DOI: 10.3390/ijms20225615] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
The etiopathogenesis of endometriosis is a multifactorial process resulting in a heterogeneous disease. Considering that endometriosis etiology and pathogenesis are still far from being fully elucidated, the current review aims to offer a comprehensive summary of the available evidence. We performed a narrative review synthesizing the findings of the English literature retrieved from computerized databases from inception to June 2019, using the Medical Subject Headings (MeSH) unique ID term "Endometriosis" (ID:D004715) with "Etiology" (ID:Q000209), "Immunology" (ID:Q000276), "Genetics" (ID:D005823) and "Epigenesis, Genetic" (ID:D044127). Endometriosis may origin from Müllerian or non-Müllerian stem cells including those from the endometrial basal layer, Müllerian remnants, bone marrow, or the peritoneum. The innate ability of endometrial stem cells to regenerate cyclically seems to play a key role, as well as the dysregulated hormonal pathways. The presence of such cells in the peritoneal cavity and what leads to the development of endometriosis is a complex process with a large number of interconnected factors, potentially both inherited and acquired. Genetic predisposition is complex and related to the combined action of several genes with limited influence. The epigenetic mechanisms control many of the processes involved in the immunologic, immunohistochemical, histological, and biological aberrations that characterize the eutopic and ectopic endometrium in affected patients. However, what triggers such alterations is not clear and may be both genetically and epigenetically inherited, or it may be acquired by the particular combination of several elements such as the persistent peritoneal menstrual reflux as well as exogenous factors. The heterogeneity of endometriosis and the different contexts in which it develops suggest that a single etiopathogenetic model is not sufficient to explain its complex pathobiology.
Collapse
Affiliation(s)
- Antonio Simone Laganà
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Piazza Biroldi 1, 21100 Varese, Italy; (S.G.); (F.G.)
| | - Simone Garzon
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Piazza Biroldi 1, 21100 Varese, Italy; (S.G.); (F.G.)
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, D-48149 Münster, Germany;
| | - Paola Viganò
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina 60, 20136 Milan, Italy;
| | - Massimo Franchi
- Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Piazzale Aristide Stefani 1, 37126 Verona, Italy;
| | - Fabio Ghezzi
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Piazza Biroldi 1, 21100 Varese, Italy; (S.G.); (F.G.)
| | - Dan C. Martin
- School of Medicine, University of Tennessee Health Science Center, 910 Madison Ave, Memphis, TN 38163, USA;
- Virginia Commonwealth University, 907 Floyd Ave, Richmond, VA 23284, USA
| |
Collapse
|
10
|
Loh CY, Chai JY, Tang TF, Wong WF, Sethi G, Shanmugam MK, Chong PP, Looi CY. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019; 8:E1118. [PMID: 31547193 PMCID: PMC6830116 DOI: 10.3390/cells8101118] [Citation(s) in RCA: 823] [Impact Index Per Article: 137.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 12/17/2022] Open
Abstract
Epithelial-to-Mesenchymal Transition (EMT) has been shown to be crucial in tumorigenesis where the EMT program enhances metastasis, chemoresistance and tumor stemness. Due to its emerging role as a pivotal driver of tumorigenesis, targeting EMT is of great therapeutic interest in counteracting metastasis and chemoresistance in cancer patients. The hallmark of EMT is the upregulation of N-cadherin followed by the downregulation of E-cadherin, and this process is regulated by a complex network of signaling pathways and transcription factors. In this review, we summarized the recent understanding of the roles of E- and N-cadherins in cancer invasion and metastasis as well as the crosstalk with other signaling pathways involved in EMT. We also highlighted a few natural compounds with potential anti-EMT property and outlined the future directions in the development of novel intervention in human cancer treatments. We have reviewed 287 published papers related to this topic and identified some of the challenges faced in translating the discovery work from bench to bedside.
Collapse
Affiliation(s)
- Chin-Yap Loh
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| | - Jian Yi Chai
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| | - Ting Fang Tang
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| | - Muthu Kumaraswamy Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| | - Pei Pei Chong
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| |
Collapse
|
11
|
Luo W, Fedda F, Lynch P, Tan D. CDH1 Gene and Hereditary Diffuse Gastric Cancer Syndrome: Molecular and Histological Alterations and Implications for Diagnosis And Treatment. Front Pharmacol 2018; 9:1421. [PMID: 30568591 PMCID: PMC6290068 DOI: 10.3389/fphar.2018.01421] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer, a group of common malignancies, results in the most cancer mortality worldwide after only lung and colorectal cancer. Although familial gastric cancers have long been recognized, it was not until recently that they were discovered to be associated with mutations of specific genes. Mutations of CDH1, the gene encoding E-cadherin, are the most common germline mutations detected in gastric cancer and underlie hereditary diffuse gastric cancer (HDGC) syndrome. All reported HDGCs are the pure diffuse type by Lauren classification and are associated with dismal prognosis once the tumor invades the submucosa. Because CDH1 germline mutations are inherited in an autosomal-dominant fashion and have high penetrance, the International Gastric Cancer Linkage Consortium (IGCLC) developed criteria to facilitate the screening of CDH1 mutation carriers; these criteria have been proven to have excellent sensitivity and specificity. Recent histologic studies suggest that HDGC progresses through several stages. Even when the tumor becomes "invasive" in lamina propria, it may stay indolent for a long time. However, the molecular mechanisms that induce the transitions between stages and determine the length of the indolent phase remain to be determined. Although the standard management for CDH1 mutation carriers is prophylactic total gastrectomy, many questions must be answered before the surgery can be done. These include the optimal surveillance strategy, the best strategy to choose surgical candidates, and the ideal time to perform surgery. In addition to increasing the risk of gastric cancer, CDH1 germline mutations also increase the risk of invasive lobular carcinoma of the breast, and possibly colorectal adenocarcinoma, and are associated with blepharocheilodontic syndrome (a congenital development disorder). However, the optimal management of these conditions is less established owing to insufficient data regarding the risk of cancer development. This review focuses on molecular and histological findings in HDGC, as opposed to sporadic diffuse gastric cancer, and their implications for the management of CDH1 mutation carriers and the diagnosis and treatment of HDGC. Other conditions associated with CDH1 germline mutations and future research directions are also discussed.
Collapse
Affiliation(s)
- Wenyi Luo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Faysal Fedda
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Patrick Lynch
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dongfeng Tan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
12
|
Fabianova A, Barathova M, Csaderova L, Simko V, Zatovicova M, Labudova M, Pastorek J. Hypoxic marker CA IX and adhesion mediator β-catenin are downregulated by lymphocytic choriomeningitis virus persistent infection. Oncotarget 2018; 9:12879-12893. [PMID: 29560117 PMCID: PMC5849181 DOI: 10.18632/oncotarget.24387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/25/2018] [Indexed: 11/25/2022] Open
Abstract
Renal cell carcinoma is one of the most frequent cancer diseases with high resistance to radio- and chemotherapy. Mutation of VHL gene is frequent in these tumors leading to simulation of hypoxic conditions. Lymphocytic choriomeningitis virus, belonging to RNA viruses, is a neglected human pathogen and teratogen. We have found that infection of renal cell carcinoma cells by lymphocytic choriomeningitis virus strain MX causes a decrease of carbonic anhydrase IX protein and RNA level. Lower expression of carbonic anhydrase IX on the cell surface provides less target for carbonic anhydrase IX-targeted immunotherapy. What more, reduced levels of adhesion mediating protein β-catenin as well as E-cadherin, as a consequence of infection, suggest a possible increase in metastatic potential of cells infected by lymphocytic choriomeningitis virus strain MX. These results might help elucidate differences in patients susceptibility to immunotherapy directed against carbonic anhydrase IX or in developing new therapeutical strategies. Our data indicate that presence of infection can significantly affect patient response to cancer therapy.
Collapse
Affiliation(s)
- Andrea Fabianova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 845 05, Slovak Republic
| | - Monika Barathova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 845 05, Slovak Republic
| | - Lucia Csaderova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 845 05, Slovak Republic
| | - Veronika Simko
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 845 05, Slovak Republic
| | - Miriam Zatovicova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 845 05, Slovak Republic
| | - Martina Labudova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 845 05, Slovak Republic
| | - Jaromir Pastorek
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 845 05, Slovak Republic.,Department of Chemistry, Faculty of Natural Sciences, University of SS. Cyril and Methodius, Trnava 917 01, Slovak Republic
| |
Collapse
|
13
|
Sun H, Liu M, Wu X, Yang C, Zhang Y, Xu Z, Gao K, Wang F. Overexpression of N-cadherin and β-catenin correlates with poor prognosis in patients with nasopharyngeal carcinoma. Oncol Lett 2017; 13:1725-1730. [PMID: 28454316 DOI: 10.3892/ol.2017.5645] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 10/27/2016] [Indexed: 12/20/2022] Open
Abstract
An increasing amount of evidence demonstrates that epithelial-mesenchymal transition (EMT) is important in tumor invasion and metastases. The cell-cell adhesion molecule N-cadherin and the Wnt/β-catenin cascade protein β-catenin are two biomarkers of EMT. The present study aimed to measure the expression levels of N-cadherin and β-catenin in samples from patients with nasopharyngeal carcinoma (NPC) and evaluate their prognostic significance. N-cadherin and β-catenin mRNA was evaluated using reverse transcription-quantitative polymerase chain reaction in 26 NPC tissue samples and 8 nasopharyngeal epithelium samples. Protein expression of N-cadherin and β-catenin was also detected using immunohistochemistry in 128 archival NPC paraffin-embedded specimens. Finally, associations between clinical pathological parameters and prognostic values in NPC were evaluated. The results demonstrated that both the mRNA and protein levels of N-cadherin and β-catenin were significantly increased in NPC tissues compared with the controls. Enhanced expression of N-cadherin and β-catenin protein was strongly correlated with the status of lymph node metastasis and clinical stages in patients with NPC. Notably, high expression of N-cadherin and β-catenin proteins was significantly correlated with lower overall survival (OS) rate in patients with NPC. Finally, multivariate analysis demonstrated that expression of N-cadherin protein and clinical stages were independent prognostic factors for patients with NPC. Therefore, the present study demonstrated that N-cadherin and β-catenin expression may be used as potential prognostic biomarkers for patients with NPC.
Collapse
Affiliation(s)
- Hong Sun
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mingyu Liu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xuewen Wu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Chunguang Yang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yanni Zhang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhenhang Xu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Kelei Gao
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Fengjun Wang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
14
|
Deregulated MicroRNAs in Biliary Tract Cancer: Functional Targets and Potential Biomarkers. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4805270. [PMID: 27957497 PMCID: PMC5120202 DOI: 10.1155/2016/4805270] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/10/2016] [Indexed: 02/07/2023]
Abstract
Biliary tract cancer (BTC) is still a fatal disease with very poor prognosis. The lack of reliable biomarkers for early diagnosis and of effective therapeutic targets is a major demanding problem in diagnosis and management of BTC. Due to the clinically silent and asymptomatic characteristics of the tumor, most patients are diagnosed at an already advanced stage allowing only for a palliative therapeutic approach. MicroRNAs are small noncoding RNAs well known to regulate various cellular functions and pathologic events including the formation and progression of cancer. Over the last years, several studies have shed light on the role of microRNAs in BTC, making them potentially attractive therapeutic targets and candidates as biomarkers. In this review, we will focus on the role of oncogenic and tumor suppressor microRNAs and their direct targets in BTC. Furthermore, we summarize and discuss data that evaluate the diagnostic power of deregulated microRNAs as possible future biomarkers for BTC.
Collapse
|
15
|
Carvalho S, Catarino TA, Dias AM, Kato M, Almeida A, Hessling B, Figueiredo J, Gärtner F, Sanches JM, Ruppert T, Miyoshi E, Pierce M, Carneiro F, Kolarich D, Seruca R, Yamaguchi Y, Taniguchi N, Reis CA, Pinho SS. Preventing E-cadherin aberrant N-glycosylation at Asn-554 improves its critical function in gastric cancer. Oncogene 2016; 35:1619-1631. [PMID: 26189796 PMCID: PMC4856288 DOI: 10.1038/onc.2015.225] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 05/15/2015] [Accepted: 05/18/2015] [Indexed: 11/09/2022]
Abstract
E-cadherin is a central molecule in the process of gastric carcinogenesis and its posttranslational modifications by N-glycosylation have been described to induce a deleterious effect on cell adhesion associated with tumor cell invasion. However, the role that site-specific glycosylation of E-cadherin has in its defective function in gastric cancer cells needs to be determined. Using transgenic mice models and human clinical samples, we demonstrated that N-acetylglucosaminyltransferase V (GnT-V)-mediated glycosylation causes an abnormal pattern of E-cadherin expression in the gastric mucosa. In vitro models further indicated that, among the four potential N-glycosylation sites of E-cadherin, Asn-554 is the key site that is selectively modified with β1,6 GlcNAc-branched N-glycans catalyzed by GnT-V. This aberrant glycan modification on this specific asparagine site of E-cadherin was demonstrated to affect its critical functions in gastric cancer cells by affecting E-cadherin cellular localization, cis-dimer formation, molecular assembly and stability of the adherens junctions and cell-cell aggregation, which was further observed in human gastric carcinomas. Interestingly, manipulating this site-specific glycosylation, by preventing Asn-554 from receiving the deleterious branched structures, either by a mutation or by silencing GnT-V, resulted in a protective effect on E-cadherin, precluding its functional dysregulation and contributing to tumor suppression.
Collapse
Affiliation(s)
- S Carvalho
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - TA Catarino
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - AM Dias
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - M Kato
- Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, Wako, Saitama, Japan
| | - A Almeida
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Berlin, Germany
| | - B Hessling
- Center for Molecular Biology, University of Heidelberg, Heidelberg, Germany
| | - J Figueiredo
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - F Gärtner
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - JM Sanches
- Institute for Systems and Robotics (ISR/IST), LARSyS, Instituto Superior Técnico, University of Lisbon, Lisboa, Portugal
| | - T Ruppert
- Center for Molecular Biology, University of Heidelberg, Heidelberg, Germany
| | - E Miyoshi
- Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan
| | - M Pierce
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - F Carneiro
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Medical Faculty, University of Porto, Porto, Portugal
- Department of Pathology, Hospital S. Joao, Porto, Portugal
| | - D Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - R Seruca
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Medical Faculty, University of Porto, Porto, Portugal
| | - Y Yamaguchi
- Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, Wako, Saitama, Japan
| | - N Taniguchi
- Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, Wako, Saitama, Japan
| | - CA Reis
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Medical Faculty, University of Porto, Porto, Portugal
| | - SS Pinho
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal/Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
16
|
Dong S, Zhao J, Wei J, Bowser RK, Khoo A, Liu Z, Luketich JD, Pennathur A, Ma H, Zhao Y. F-box protein complex FBXL19 regulates TGFβ1-induced E-cadherin down-regulation by mediating Rac3 ubiquitination and degradation. Mol Cancer 2014; 13:76. [PMID: 24684802 PMCID: PMC3994216 DOI: 10.1186/1476-4598-13-76] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 03/25/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Rac3 is a small GTPase multifunctional protein that regulates cell adhesion, migration, and differentiation. It has been considered as an oncogene in breast cancer; however, its role in esophageal cancer and the regulation of its stability have not been studied. F-box proteins are major subunits within the Skp1-Cullin-1-F-box (SCF) E3 ubiquitin ligases that recognize particular substrates for ubiquitination and proteasomal degradation. Recently, we have shown that SCFFBXL19 targets Rac1 and RhoA, thus regulating Rac1 and RhoA ubiquitination and degradation. Here, we demonstrate the role of FBXL19 in the regulation of Rac3 site-specific ubiquitination and stability. Expression of TGFβ1 is associated with poor prognosis of esophageal cancer. TGFβ1 reduces tumor suppressor, E-cadherin, expression in various epithelial-derived cancers. Here we investigate the role of FBXL19-mediated Rac3 degradation in TGFβ1-induced E-cadherin down-regulation in esophageal cancer cells. METHODS FBXL19-regulated endogenous and over-expressed Rac3 stability were determined by immunoblotting and co-immunoprecipitation. Esophageal cancer cells (OE19 and OE33) were used to investigate TGFβ1-induced E-cadherin down-regulation by Immunoblotting and Immunostaining. RESULTS Overexpression of FBXL19 decreased endogenous and over-expressed Rac3 expression by interacting and polyubiquitinating Rac3, while down-regulation of FBXL19 suppressed Rac3 degradation. Lysine166 within Rac3 was identified as an ubiquitination acceptor site. The FBXL19 variant with truncation at the N-terminus resulted in an increase in Rac3 degradation; however, the FBXL19 variant with truncation at the C-terminus lost its ability to interact with Rac3 and ubiquitinate Rac3 protein. Further, we found that Rac3 plays a critical role in TGFβ1-induced E-cadherin down-regulation in esophageal cancer cells. Over-expression of FBXL19 attenuated TGFβ1-induced E-cadherin down-regulation and esophageal cancer cells elongation phenotype. CONCLUSIONS Collectively these data unveil that FBXL19 functions as an antagonist of Rac3 by regulating its stability and regulates the TGFβ1-induced E-cadherin down-regulation. This study will provide a new potential therapeutic strategy to regulate TGFβ1 signaling, thus suppressing esophageal tumorigenesis.
Collapse
Affiliation(s)
- Su Dong
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun, China
- Department of Anesthesia, First Hospital of Jilin University, Changchun, China
- Department of Medicine and the Acute Lung Injury Center of Excellence, University of Pittsburgh, 3459 Fifth Avenue, NW 628MUH, Pittsburgh 15213, PA, USA
| | - Jing Zhao
- Department of Medicine and the Acute Lung Injury Center of Excellence, University of Pittsburgh, 3459 Fifth Avenue, NW 628MUH, Pittsburgh 15213, PA, USA
| | - Jianxin Wei
- Department of Medicine and the Acute Lung Injury Center of Excellence, University of Pittsburgh, 3459 Fifth Avenue, NW 628MUH, Pittsburgh 15213, PA, USA
| | - Rachel K Bowser
- Department of Medicine and the Acute Lung Injury Center of Excellence, University of Pittsburgh, 3459 Fifth Avenue, NW 628MUH, Pittsburgh 15213, PA, USA
| | - Andrew Khoo
- Department of Medicine and the Acute Lung Injury Center of Excellence, University of Pittsburgh, 3459 Fifth Avenue, NW 628MUH, Pittsburgh 15213, PA, USA
| | - Zhonghui Liu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - James D Luketich
- Department of Thoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Arjun Pennathur
- Department of Thoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haichun Ma
- Department of Anesthesia, First Hospital of Jilin University, Changchun, China
| | - Yutong Zhao
- Department of Medicine and the Acute Lung Injury Center of Excellence, University of Pittsburgh, 3459 Fifth Avenue, NW 628MUH, Pittsburgh 15213, PA, USA
| |
Collapse
|
17
|
Vasconcelos-Nóbrega C, Costa C, Vala H, Colaço A, Santos L, Lopes C, Oliveira PA. E-cadherin and β-catenin expression during urothelial carcinogenesis induced by N-butyl-N-(4-hydroxybutyl) nitrosamine in mice. Urol Int 2013; 91:462-6. [PMID: 23548313 DOI: 10.1159/000348329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 01/17/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND E-cadherin and β-catenin are adhesion molecules that promote integrity and stability of the urothelium. A decrease in their expression is associated with more aggressive tumour phenotypes with the ability to invade and metastasize. MATERIAL AND METHODS 45 ICR male mice were used, of which 25 received N-butyl-N-(4-hydroxybutyl)nitrosamine (0.05%) in drinking water for a period of 12 weeks. Immunohistochemical expression was evaluated in all urinary bladder preparations for E-cadherin and for β-catenin. RESULTS Preneoplastic lesions showed staining patterns similar to normal urothelium. In simple and nodular hyperplasia, membrane staining was dominant (66.7-78.6 and 50-100%, respectively). In dysplasia a cytoplasmic pattern was prevalent (86.7-100%). Neoplastic lesions exhibit an abnormal staining pattern (100%) with heterogeneous staining (cytoplasmic, nuclear and membrane staining). A strong correlation was observed between both adhesion molecule staining patterns (r = 0.83; p = 0.039). CONCLUSIONS In mice, as in humans, E-cadherin and β-catenin are valuable tools to investigate cellular adhesion status of urothelium and can be considered as indicators of tumour aggressiveness and evolution.
Collapse
|
18
|
Bourboulia D, Han H, Jensen-Taubman S, Gavil N, Isaac B, Wei B, Neckers L, Stetler-Stevenson WG. TIMP-2 modulates cancer cell transcriptional profile and enhances E-cadherin/beta-catenin complex expression in A549 lung cancer cells. Oncotarget 2013; 4:166-76. [PMID: 23371049 PMCID: PMC3702216 DOI: 10.18632/oncotarget.801] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 01/26/2013] [Indexed: 12/22/2022] Open
Abstract
Tissue Inhibitor of Metalloproteinase 2 (TIMP-2) plays an essential role in regulating matrix remodeling, cell growth, differentiation, angiogenesis and apoptosis in vitro and in vivo. We have recently shown that TIMP-2-mediated inhibition of tumor growth is independent of matrix metalloproteinase-mediated mechanisms, and is a consequence of modulating both the tumor cells and the tumor microenvironment. In the current study we aim to identify the molecular pathways associated with these effects. We analyzed the transcriptional profile of the human lung cancer cell line A549 upon overexpression of TIMP-2 and Ala+TIMP-2 (mutant that does not inhibit MMP activity), and we found changes in gene expression predominantly related to decreased tumor development and metastasis. Increased E-cadherin expression in response to both TIMP-2 and Ala+TIMP-2 expression was confirmed by real time quantitative RT-PCR and immunoblotting. A549 cells treated with epidermal growth factor (EGF) displayed loss of cobblestone morphology and cell-cell contact, while cells overexpressing TIMP-2 or Ala+TIMP-2 were resistant to EGF-induced morphological changes. Moreover, exogenous treatment with recombinant Ala+TIMP-2 blocked EGF induced down-regulation of E-cadherin. In vivo, immunohistochemistry of A549 xenografts expressing either TIMP-2 or Ala+TIMP-2 demonstrated increased E-cadherin protein levels. More importantly, transcriptional profile analysis of tumor tissue revealed critical pathways associated with effects on tumor-host interaction and inhibition of tumor growth. In conclusion, we show that TIMP-2 promotes an anti-tumoral transcriptional profile in vitro and in vivo, including upregulation of E-cadherin, in A549 lung cancer cells.
Collapse
Affiliation(s)
- Dimitra Bourboulia
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Advanced Technology Center, 8717 Grovemont Circle, Bethesda, MD, USA
| | - HuiYing Han
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Advanced Technology Center, 8717 Grovemont Circle, Bethesda, MD, USA
| | - Sandra Jensen-Taubman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Advanced Technology Center, 8717 Grovemont Circle, Bethesda, MD, USA
| | - Noah Gavil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Advanced Technology Center, 8717 Grovemont Circle, Bethesda, MD, USA
- Bowdoin College, Brunswick, ME, USA
| | - Biju Isaac
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Advanced Technology Center, 8717 Grovemont Circle, Bethesda, MD, USA
- Center for Computational Science, University of Miami, Miami, FL, USA
| | - Beiyang Wei
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Advanced Technology Center, 8717 Grovemont Circle, Bethesda, MD, USA
| | - Len Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - William G. Stetler-Stevenson
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Advanced Technology Center, 8717 Grovemont Circle, Bethesda, MD, USA
| |
Collapse
|
19
|
Yang L, Xu L. GPR56 in cancer progression: current status and future perspective. Future Oncol 2012; 8:431-40. [PMID: 22515446 DOI: 10.2217/fon.12.27] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cell adhesion is a critical process during cancer progression and is mediated by transmembrane receptors. Recently, GPR56, a member of the adhesion family of G protein-coupled receptors, was established as a new type of adhesion receptor that binds to extracellular matrix proteins and shown to play inhibitory roles in melanoma progression. Further studies revealed that the extracellular portion and the seven transmembrane domains of GPR56 function antagonistically to regulate VEGF production and angiogenesis via a signaling pathway mediated by PKCα. Tissue transglutaminase was identified as the first extracellular matrix protein that binds to GPR56. It is a crosslinking enzyme in the extracellular matrix but is also expressed in the cytosol. Tissue transglutaminase plays pleiotropic roles in cancer progression. Whether and how it might mediate GPR56-regulated cancer progression awaits further investigation.
Collapse
Affiliation(s)
- Liquan Yang
- Department of Biomedical Genetics, Department of Dermatology, James P Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | |
Collapse
|
20
|
Kreiseder B, Orel L, Bujnow C, Buschek S, Pflueger M, Schuett W, Hundsberger H, de Martin R, Wiesner C. α-Catulin downregulates E-cadherin and promotes melanoma progression and invasion. Int J Cancer 2012; 132:521-30. [PMID: 22733455 DOI: 10.1002/ijc.27698] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Accepted: 06/06/2012] [Indexed: 11/07/2022]
Abstract
Metastasis is associated with poor prognosis for melanoma responsible for about 90% of skin cancer-related mortality. To metastasize, melanoma cells must escape keratinocyte control, invade across the basement membrane and survive in the dermis by resisting apoptosis before they can intravasate into the circulation. α-Catulin (CTNNAL1) is a cytoplasmic molecule that integrates the crosstalk between nuclear factor-kappa B and Rho signaling pathways, binds to β-catenin and increases the level of both α-catenin and β-catenin and therefore has potential effects on inflammation, apoptosis and cytoskeletal reorganization. Here, we show that α-catulin is highly expressed in melanoma cells. Expression of α-catulin promoted melanoma progression and occurred concomitantly with the downregulation of E-cadherin and the upregulation of expression of mesenchymal genes such as N-cadherin, Snail/Slug and the matrix metalloproteinases 2 and 9. Knockdown of α-catulin promoted adhesion to and inhibited migration away from keratinocytes in an E-cadherin-dependent manner and decreased the transmigration through a keratinocyte monolayer, as well as in Transwell assays using collagens, laminin and fibronectin coating. Moreover, knockdown promoted homotypic spheroid formation and concomitantly increased E-cadherin expression along with downregulation of transcription factors implicated in its repression (Snail/Slug, Twist and ZEB). Consistent with the molecular changes, α-catulin provoked invasion of melanoma cells in a three-dimensional culture assay by the upregulation of matrix metalloproteinases 2 and 9 and the activation of ROCK/Rho. As such, α-catulin may represent a key driver of the metastatic process, implicating potential for therapeutic interference.
Collapse
Affiliation(s)
- Birgit Kreiseder
- Medical and Pharmaceutical Biotechnology, University of Applied Sciences, Krems, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Expression of E-cadherin and α-catenin in a varicocele-induced infertility rat model. Asian J Androl 2011; 13:470-5. [PMID: 21399649 DOI: 10.1038/aja.2010.94] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The roles of E-cadherin and α-catenin were evaluated in the development of varicocele-induced infertility. Analysis of the association between the expression of E-cadherin/α-catenin and clinical/pathological parameters was performed. Thirty 10-week-old male rats (experimental group) were used for the experiments; the left renal vein was ligated to form a varicocele. The abdomen was incised in 30 rats (control group) and no procedure was performed on 10 rats (baseline group). The weights of the left testis, serum reactive oxygen species (ROS), testosterone, luteinizing hormone (LH), follicle-stimulating hormone (FSH) and degenerative changes in the seminiferous tubules after 4 and 8 weeks were recorded. The expression of E-cadherin and α-catenin was evaluated by immunohistochemical (IHC) staining and Western blot analysis. The ROS increased in the 8-week experimental group, compared with the baseline and control groups (P < 0.001 for both). Additionally, FSH significantly increased in the 4- and 8-week experimental group compared with the control groups (P = 0.013 and P = 0.032, respectively). The ratio of degenerative changes in the seminiferous tubules of the experimental groups increased. The IHC staining showed that the expression of E-cadherin and α-catenin decreased in the 4- and 8-week experimental groups. Similar to the IHC staining, the experimental group had decreased reactivity on Western blot analysis. The expression of E-cadherin and α-catenin was significantly associated with the ROS and degenerative changes in the seminiferous tubules. The results of this study suggest that damage to the blood-testis barrier (BTB) is associated with varicocele-induced male infertility, and that ROS may cause damage to the BTB.
Collapse
|
22
|
Zeckey C, Dahm M, Wallrath A, Herr M, Walter Kunz H, Vahl CF, Horn S. Synthetic (glyco-)peptides of the homophilic recognition domain of E-cadherin lead to increased E-cadherin mRNA synthesis and are inductors of cell differentiation in primary lung cancer cell lines. Pathol Res Pract 2010; 206:450-7. [PMID: 20403671 DOI: 10.1016/j.prp.2010.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 02/12/2010] [Indexed: 10/19/2022]
Abstract
E-cadherin is one of the critical molecules involved in the metastatic process in many types of cancer. Once combined, E-cadherin exceeds the amount of membranous E-cadherin on the cellular surface by activation of intracellular signaling cascades. Studies on transformed keratinocytes of the HaCat cell line showed induction of differentiation by synthetical partial structures of the homophilic binding region of E-cadherin. The knowledge of effects in lung cancer cells is sparse. Therefore, the effects in primary lung cancer cell lines were investigated. Four primary lung cancer cell lines were incubated for 3, 6, 12, 15, 18, and 24h with synthetic partial structures (peptide and glycopeptide). The control substance was sodium butyrate. mRNA was isolated, and relative quantification of E-cadherin was performed using the Real-Time PCR. During the stimulation period, morphologic pictures were taken, and immunohistochemical staining of membranous E-cadherin was performed. Life/dead assays were used to display cell vitality. The intracellular E-cadherin mRNA amount was increased after incubation with the synthetic partial structures. Life/dead assays showed improved survival and integrated cell/cell bindings after stimulation with the partial structures. Increased cell mortality was revealed after sodium butyrate incubation. An effect mediated via E-cadherin on the cellular surface is proposed. The two synthetic partial structures of the homophilic binding region of E-cadherin increased the intracellular E-cadherin mRNA amount, cell-cell bindings, and survival of the tumor cells. Extracellular binding by synthetic partial structures to the binding region may have a beneficial influence on tumor progression in the metastatic process.
Collapse
Affiliation(s)
- Christian Zeckey
- Department of Cardio-Thoracic and Vascular Surgery, University Medical Center of the Johannes Gutenberg University Mainz, Germany.
| | | | | | | | | | | | | |
Collapse
|
23
|
Cannito S, Novo E, di Bonzo LV, Busletta C, Colombatto S, Parola M. Epithelial-mesenchymal transition: from molecular mechanisms, redox regulation to implications in human health and disease. Antioxid Redox Signal 2010; 12:1383-430. [PMID: 19903090 DOI: 10.1089/ars.2009.2737] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a fundamental process, paradigmatic of the concept of cell plasticity, that leads epithelial cells to lose their polarization and specialized junctional structures, to undergo cytoskeleton reorganization, and to acquire morphological and functional features of mesenchymal-like cells. Although EMT has been originally described in embryonic development, where cell migration and tissue remodeling have a primary role in regulating morphogenesis in multicellular organisms, recent literature has provided evidence suggesting that the EMT process is a more general biological process that is also involved in several pathophysiological conditions, including cancer progression and organ fibrosis. This review offers first a comprehensive introduction to describe major relevant features of EMT, followed by sections dedicated on those signaling mechanisms that are known to regulate or affect the process, including the recently proposed role for oxidative stress and reactive oxygen species (ROS). Current literature data involving EMT in both physiological conditions (i.e., embryogenesis) and major human diseases are then critically analyzed, with a special final focus on the emerging role of hypoxia as a relevant independent condition able to trigger EMT.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Experimental Medicine and Oncology and Interuniversity Center for Hepatic Pathophysiology, University of Turin, Turin, Italy
| | | | | | | | | | | |
Collapse
|
24
|
Xu C, Lo A, Yammanuru A, Tallarico ASC, Brady K, Murakami A, Barteneva N, Zhu Q, Marasco WA. Unique biological properties of catalytic domain directed human anti-CAIX antibodies discovered through phage-display technology. PLoS One 2010; 5:e9625. [PMID: 20224781 PMCID: PMC2835754 DOI: 10.1371/journal.pone.0009625] [Citation(s) in RCA: 34] [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: 12/24/2009] [Accepted: 02/05/2010] [Indexed: 11/18/2022] Open
Abstract
Carbonic anhydrase IX (CAIX, gene G250/MN-encoded transmembrane protein) is highly expressed in various human epithelial tumors such as renal clear cell carcinoma (RCC), but absent from the corresponding normal tissues. Besides the CA signal transduction activity, CAIX may serve as a biomarker in early stages of oncogenesis and also as a reliable marker of hypoxia, which is associated with tumor resistance to chemotherapy and radiotherapy. Although results from preclinical and clinical studies have shown CAIX as a promising target for detection and therapy for RCC, only a limited number of murine monoclonal antibodies (mAbs) and one humanized mAb are available for clinical testing and development. In this study, paramagnetic proteoliposomes of CAIX (CAIX-PMPLs) were constructed and used for anti-CAIX antibody selection from our 27 billion human single-chain antibody (scFv) phage display libraries. A panel of thirteen human scFvs that specifically recognize CAIX expressed on cell surface was identified, epitope mapped primarily to the CA domain, and affinity-binding constants (KD) determined. These human anti-CAIX mAbs are diverse in their functions including induction of surface CAIX internalization into endosomes and inhibition of the carbonic anhydrase activity, the latter being a unique feature that has not been previously reported for anti-CAIX antibodies. These human anti-CAIX antibodies are important reagents for development of new immunotherapies and diagnostic tools for RCC treatment as well as extending our knowledge on the basic structure-function relationships of the CAIX molecule.
Collapse
Affiliation(s)
- Chen Xu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Agnes Lo
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anuradha Yammanuru
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aimee St. Clair Tallarico
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kristen Brady
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Akikazu Murakami
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Natasha Barteneva
- Immune Disease Institute and Program in Cellular and Molecular Medicine, Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Quan Zhu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (QZ); (WAM)
| | - Wayne A. Marasco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (QZ); (WAM)
| |
Collapse
|
25
|
ADAM10 Is Upregulated in Melanoma Metastasis Compared with Primary Melanoma. J Invest Dermatol 2010; 130:763-73. [DOI: 10.1038/jid.2009.335] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Sarrió D, Palacios J, Hergueta-Redondo M, Gómez-López G, Cano A, Moreno-Bueno G. Functional characterization of E- and P-cadherin in invasive breast cancer cells. BMC Cancer 2009; 9:74. [PMID: 19257890 PMCID: PMC2656544 DOI: 10.1186/1471-2407-9-74] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Accepted: 03/03/2009] [Indexed: 12/18/2022] Open
Abstract
Background Alterations in the cadherin-catenin adhesion complexes are involved in tumor initiation, progression and metastasis. However, the functional implication of distinct cadherin types in breast cancer biology is still poorly understood. Methods To compare the functional role of E-cadherin and P-cadherin in invasive breast cancer, we stably transfected these molecules into the MDA-MB-231 cell line, and investigated their effects on motility, invasion and gene expression regulation. Results Expression of either E- and P-cadherin significantly increased cell aggregation and induced a switch from fibroblastic to epithelial morphology. Although expression of these cadherins did not completely reverse the mesenchymal phenotype of MDA-MB-231 cells, both E- and P-cadherin decreased fibroblast-like migration and invasion through extracellular matrix in a similar way. Moreover, microarray gene expression analysis of MDA-MB-231 cells after expression of E- and P-cadherins revealed that these molecules can activate signaling pathways leading to significant changes in gene expression. Although the expression patterns induced by E- and P-cadherin showed more similarities than differences, 40 genes were differentially modified by the expression of either cadherin type. Conclusion E- and P-cadherin have similar functional consequences on the phenotype and invasive behavior of MDA-MB-231 cells. Moreover, we demonstrate for the first time that these cadherins can induce both common and specific gene expression programs on invasive breast cancer cells. Importantly, these identified genes are potential targets for future studies on the functional consequences of altered cadherin expression in human breast cancer.
Collapse
Affiliation(s)
- David Sarrió
- Department of Biochemistry UAM, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Loss of the coxsackie and adenovirus receptor (CAR) has previously been observed in gastric cancer. The role of CAR in gastric cancer pathobiology, however, is unclear. We therefore analysed CAR in 196 R0-resected gastric adenocarcinomas and non-cancerous gastric mucosa samples using immunohistochemistry and immunofluorescence. Coxsackie and adenovirus receptor was found at the surface and foveolar epithelium of all non-neoplastic gastric mucosa samples (n=175), whereas only 56% of gastric cancer specimens showed CAR positivity (P<0.0001). Loss of CAR correlated significantly with decreased differentiation, increased infiltrative depths, presence of distant metastases, and was also associated with reduced carcinoma-specific survival. To clarify whether CAR impacts the tumorbiologic properties of gastric cancer, we subsequently determined the role of CAR in proliferation, migration, and invasion of gastric cancer cell lines by application of specific CAR siRNA or ectopic expression of a human full-length CAR cDNA. These experiments showed that RNAi-mediated CAR knock down resulted in increased proliferation, migration, and invasion of gastric cancer cell lines, whereas enforced ectopic CAR expression led to opposite effects. We conclude that the association of reduced presence of CAR in more severe disease states, together with our findings in gastric cancer cell lines, suggests that CAR functionally contributes to gastric cancer pathogenesis, showing features of a tumour suppressor.
Collapse
|
28
|
Baum B, Settleman J, Quinlan MP. Transitions between epithelial and mesenchymal states in development and disease. Semin Cell Dev Biol 2008; 19:294-308. [PMID: 18343170 DOI: 10.1016/j.semcdb.2008.02.001] [Citation(s) in RCA: 298] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 02/04/2008] [Indexed: 12/21/2022]
Abstract
The ancestors of modern Metazoa were constructed in large part by the foldings and distortions of two-dimensional sheets of epithelial cells. This changed approximately 600 million years ago with the evolution of mesenchymal cells. These cells arise as the result of epithelial cell delamination through a reprogramming process called an epithelial to mesenchymal transition (EMT) [Shook D, Keller R. Mechanisms, mechanics and function of epithelial-mesenchymal transitions in early development. Mech Dev 2003;120:1351-83; Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 2006;7:131-42]. Because mesenchymal cells are free to migrate through the body cavity, the evolution of the mesenchyme opened up new avenues for morphological plasticity, as cells evolved the ability to take up new positions within the embryo and to participate in novel cell-cell interactions; forming new types of internal tissues and organs such as muscle and bone [Thiery JP, Sleeman, JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 2006;7:131-42; Hay ED, Zuk A. Transformations between epithelium and mesenchyme: normal, pathological, and experimentally induced. Am J Kidney Dis 1995;26:678-90]. After migrating to a suitable site, mesenchymal cells coalesce and re-polarize to form secondary epithelia, in a so-called mesenchymal-epithelial transition (MET). Such switches between mesenchymal and epithelial states are a frequent feature of Metazoan gastrulation [Hay ED, Zuk A. Transformations between epithelium and mesenchyme: normal, pathological, and experimentally induced. Am J Kidney Dis 1995;26:678-90] and the neural crest lineage [Duband JL, Monier F, Delannet M, Newgreen D. Epitheliu-mmesenchyme transition during neural crest development. Acta Anat 1995;154:63-78]. Significantly, however, when hijacked during the development of cancer, the ability of cells to undergo EMT, to leave the primary tumor and to undergo MET at secondary sites can have devastating consequences on the organism, allowing tumor cells derived from epithelia to invade surrounding tissues and spread through the host [Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 2006;7:131-42; Hay ED, Zuk A. Transformations between epithelium and mesenchyme: normal, pathological, and experimentally induced. Am J Kidney Dis 1995;26:678-90]. Thus, the molecular and cellular mechanisms underpinning EMT are both an essential feature of Metazoan development and an important area of biomedical research. In this review, we discuss the common molecular and cellular mechanisms involved in EMT in both cases.
Collapse
Affiliation(s)
- Buzz Baum
- Department of Cell and Developmental Biology, UCL, London, UK
| | | | | |
Collapse
|
29
|
Miyashita Y, Ozawa M. A dileucine motif in its cytoplasmic domain directs β-catenin-uncoupled E-cadherin to the lysosome. J Cell Sci 2007; 120:4395-406. [DOI: 10.1242/jcs.03489] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The E-cadherin-catenin complex regulates Ca2+-dependent cell-cell adhesion and is localized to the basolateral membrane of polarized epithelial cells. Uncoupling β-catenin from E-cadherin by deletion or substitution mutations causes accumulation of these proteins in intracellular compartments, including the trans-Golgi network and early endosomes, and degradation in lysosomes. Expression of a dominant-negative dynamin did not change the pattern of the mutant E-cadherin localization, indicating that the endocytosis of the protein from the cell surface does not contribute significantly to the accumulation of the protein in the intracellular compartments. Alternatively, E-cadherin lacking its entire cytoplasmic domain (tail-less E-cadherin) was detected on the surface of cells and targeted to the basolateral membrane. We found that 20 amino acid residues within the juxtamembrane region contain the signal responsible for intracellular accumulation and the lysosomal targeting of E-cadherin. A dileucine motif within this region seems crucial, because substitution of these residues to alanines resulted in efficient surface expression of the protein. The tail-less E-cadherin construct and the dileucine-substitution construct were detected on the basolateral membranes. Thus, the dileucine motif of E-cadherin is not required for its basolateral targeting.
Collapse
Affiliation(s)
- Yayoi Miyashita
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
30
|
Zhao YF, Zhang YG, Tian XX. Aberrant methylation of multiple genes in gastric carcinomas. Int J Surg Pathol 2007; 15:242-51. [PMID: 17652530 DOI: 10.1177/1066896907302117] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In this study, we examined aberrant methylation of the E-cadherin, estrogen receptor, RB1 , p16, p15, p14, and MGMT genes by the methylation-specific polymerase chain reaction method in 101 gastric carcinomas. Hypermethylation was detected in E-cadherin, estrogen receptor, RB1, p16, p14, p15, and MGMT at the rates of 27.7%, 44.6%, 44.6%, 30.7%, 19.2%, 7.7%, and 6.9%, respectively. A total of 82.2% cases had methylation in at least 1 of these genes, and 44.6% had methylation in 2 or more of these genes. Methylation of RB1 was associated with absence of lymph node metastasis. Methylation of estrogen receptor was associated with age and tumor location. Methylation of E-cadherin coincided with methylation of p16 or estrogen receptor. Moreover, loss of p16 protein was strongly associated with its gene methylation. This study indicates that aberrant methylation of multiple genes is involved in gastric carcinogenesis.
Collapse
Affiliation(s)
- Ying-Fang Zhao
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | | | | |
Collapse
|
31
|
Li G, Satyamoorthy K, Herlyn M. Dynamics of cell interactions and communications during melanoma development. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2007; 13:62-70. [PMID: 12097238 DOI: 10.1177/154411130201300107] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Melanoma development not only involves genetic and epigenetic changes that take place within the cell, but also involves processes determined collectively by micro-environmental factors, including cell-cell interactions and communications. During the transition from normal cells to benign and malignant lesions, and subsequently to metastatic cancer, stepwise changes in intercellular communications provide tumor cells with the ability to overcome cell-cell adhesion and micro-environmental controls from the host and to invade surrounding tissues and disperse to distant locations. Cadherins are major cell-cell adhesion molecules involved in the development and maintenance of skin. E-cadherin expressed in normal melanocytes mediates growth and invasion control by keratinocytes. Progressive loss of E-cadherin and gain of N-cadherin during melanoma development not only free melanoma cells from control by keratinocytes, but also provide new adhesion properties, resulting in switched partnerships with fibroblasts and vascular endothelial cells. The cadherin subtype switching also dictates gap junctional specificity in melanocytic cells during tumor development. This selective intercellular communication may contribute to the regulation of cell growth, differentiation, apoptosis, and migration of melanocytic cells in both physiologic and pathologic conditions. Abnormal up-regulation of the immunoglobin repeat-containing cell adhesion molecules Mel-CAM and L1-CAM potentiates invasion and migration of melanoma. Thus, abnormal expression of intercellular adhesion receptors and dysregulated intercellular communication underlies melanoma development and progression.
Collapse
Affiliation(s)
- G Li
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
32
|
Pećina-Slaus N, Zigmund M, Kusec V, Martić TN, Cacić M, Slaus M. E-cadherin and beta-catenin expression patterns in malignant melanoma assessed by image analysis. J Cutan Pathol 2007; 34:239-46. [PMID: 17302608 DOI: 10.1111/j.1600-0560.2006.00601.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND We investigated the expression of E-cadherin and beta-catenin in melanoma. Both proteins are components of adherens junctions but also play signalling roles in the wnt signal transduction pathway. MATERIALS AND METHODS Seventy malignant melanomas were analysed by immunohistochemistry and evaluated by image analysis as staining density, i.e. light permeability (LP). RESULTS Comparison of mean values of relative LP for E-cadherin and beta-catenin in tumor tissue shows that levels of E-cadherin protein are significantly lower (259.67-116.23; t=22.7; p=0.000). The comparison of mean values of the relative LP of E-cadherin in melanoma to the LP in the adjacent normal skin also shows that the expression of E-cadherin in tumor is significantly lower (256.06-169.87; t=11.55, p=0.000). beta-catenin was observed in the cytoplasm in 30.6% of patients, in 24.2% in the cell membrane, in 21% in both the cytoplasm and membrane, in 1.6% in the membrane and nucleus and in 4.8% in the cytoplasm and nucleus, whereas in 17.7% of patients beta-catenin could not be observed. Patients with Clark 4 and 5 had significantly less beta-catenin than patients with Clark 2 and 3 (chi2=12.854; p=0.005). CONCLUSIONS Changes in E-cadherin and beta-catenin levels have important roles in melanoma and could be used as molecular markers of disease progression.
Collapse
Affiliation(s)
- Nives Pećina-Slaus
- Department of Biology, Croatian Institute for brain research, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | | | | | | | | | | |
Collapse
|
33
|
Lioni M, Brafford P, Andl C, Rustgi A, El-Deiry W, Herlyn M, Smalley KSM. Dysregulation of claudin-7 leads to loss of E-cadherin expression and the increased invasion of esophageal squamous cell carcinoma cells. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:709-21. [PMID: 17255337 PMCID: PMC1851859 DOI: 10.2353/ajpath.2007.060343] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The claudins constitute a 24-member family of proteins that are critical for the function and formation of tight junctions. Here, we examine the expression of claudin-7 in squamous cell carcinoma (SCC) of the esophagus and its possible role in tumor progression. In the normal esophagus, expression of claudin-7 was confined to the cell membrane of differentiated keratinocytes. However, in the tumor samples, claudin-7 expression is often lost or localized to the cytoplasm. Assaying esophageal SCC lines revealed variable expression of claudin-7, with some lacking expression completely. Knockdown of claudin-7 in SCC cell lines using a small interfering RNA approach led to decreased E-cadherin expression, increased cell growth, and enhanced invasion into a three-dimensional matrix. The opposite was observed when claudin-7 was overexpressed in esophageal SCC cells lacking both claudin-7 and E-cadherin. In this context, the claudin-7-overexpressing cells became more adhesive and less invasive associated with increased E-cadherin expression. In summary, we demonstrate that claudin-7 is mislocalized during the malignant transformation of esophageal keratinocytes. We also demonstrate a critical role for claudin-7 expression in the regulation of E-cadherin in these cells, suggesting this may be one mechanism for the loss of epithelial architecture and invasion observed in esophageal SCC.
Collapse
Affiliation(s)
- Mercedes Lioni
- The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Mayo C, Lloreta J, Real FX, Mayol X. In vitro differentiation of HT-29 M6 mucus-secreting colon cancer cells involves a trychostatin A and p27KIP1-inducible transcriptional program of gene expression. J Cell Physiol 2007; 212:42-50. [PMID: 17311291 DOI: 10.1002/jcp.20999] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumor cell dedifferentiation-such as the loss of cell-to-cell adhesion in epithelial tumors-is associated with tumor progression. To better understand the mechanisms that maintain carcinoma cells in a differentiated state, we have dissected in vitro differentiation pathways in the mucus-secretor HT-29 M6 colon cancer cell line, which spontaneously differentiates in postconfluent cultures. By lowering the extracellular calcium concentration to levels that prevent intercellular adhesion and epithelial polarization, our results reveal that differentiation is calcium-dependent and involves: (i) a process of cell cycle exit to G(0) and (ii) the induction of a transcriptional program of differentiation gene expression (i.e., mucins MUC1 and MUC5AC, and the apical membrane peptidase DPPIV). In calcium-deprived, non-differentiated postconfluent cultures, differentiation gene promoters are repressed by a trichostatin A (TSA)-sensitive mechanism, indicating that loss of gene expression by dedifferentiation is driven by histone deacetylases (HDAC). Since TSA treatment or extracellular calcium restoration allow gene promoter activation to similar levels, we suggest that induction of differentiation is one mechanism of HDAC inhibitor antitumor action. Moreover, transcriptional de-repression can also be induced in non-differentiating culture conditions by overexpressing the cyclin-dependent kinase inhibitor p27(KIP1), which is normally induced during spontaneous differentiation. Since p27(KIP1) downregulation in colon cancer is associated with poor prognosis independently of tumor cell division rates, we propose that p27 (KIP1) may prevent tumor progression by, at least in part, enhancing the expression of some differentiation genes. Therefore, the HT-29 M6 model allows the identification of some basic mechanisms of cancer cell differentiation control, so far revealing HDAC and p27(KIP1) as key regulatory factors of differentiation gene expression.
Collapse
Affiliation(s)
- Clara Mayo
- Unitat de Biologia Celñlular i Molecular, Institut Municipal d'Investigació Mèdica, Barcelona, Spain
| | | | | | | |
Collapse
|
35
|
Pyo SW, Hashimoto M, Kim YS, Kim CH, Lee SH, Johnson KR, Wheelock MJ, Park JU. Expression of E-cadherin, P-cadherin and N-cadherin in oral squamous cell carcinoma: correlation with the clinicopathologic features and patient outcome. J Craniomaxillofac Surg 2007; 35:1-9. [PMID: 17296306 DOI: 10.1016/j.jcms.2006.11.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Accepted: 11/01/2006] [Indexed: 10/23/2022] Open
Abstract
PURPOSE Alteration of cadherin expression is associated with the loss of cellular differentiation, the acquisition of an invasive phenotype and a poor prognosis in many types of cancer. This study aimed to evaluate the immunoreactivity of E-, P- and N-cadherins (cad) in oral squamous cell carcinoma and to correlate their expression with clinicopathological features and clinical outcome. The interaction between the cadherins was also investigated. METHODS A total of 71 tissue samples were examined by immunohistochemical methods on paraffin sections using specific antibodies. RESULTS In the primary lesions and lymph node metastases, the immunoreactivity of E-cad was reduced and P-cad was over-expressed, but the expression of N-cad was negative (p<0.001, 0.01 and 0.05, respectively). The reduced primary E-cad expression was related to the invasion pattern and lymph node metastasis (p=0.046 and 0.037, respectively). However, the expression of cadherins did not appear to differ significantly in relation to the histological grade, invasion, tumour size, stage of oral SCC or tumour recurrence. A much greater reduction in the expression of E-cad was found in the positive N-cadherin group (p=0.008). Nonetheless, cadherin expression was not significantly associated with failure-free survival or overall survival in this experiment subset. CONCLUSION The reduced E-cad expression and the aberrant N-cad expression are closely associated with each other in oral cancer cases, and this suggests that cadherin switching from E. cad to N. cad may play a critical role in cancer development and metastasis.
Collapse
Affiliation(s)
- Sung Woon Pyo
- Department of Oral and Maxillofacial Surgery and Dentistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Maeda M, Johnson E, Mandal SH, Lawson KR, Keim SA, Svoboda RA, Caplan S, Wahl JK, Wheelock MJ, Johnson KR. Expression of inappropriate cadherins by epithelial tumor cells promotes endocytosis and degradation of E-cadherin via competition for p120(ctn). Oncogene 2006; 25:4595-604. [PMID: 16786001 DOI: 10.1038/sj.onc.1209396] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cadherin cell-cell adhesion proteins play an important role in modulating the behavior of tumor cells. E-cadherin serves as a suppressor of tumor cell invasion, and when tumor cells turn on the expression of a non-epithelial cadherin, they often express less E-cadherin, enhancing the tumorigenic phenotype of the cells. Here, we show that when A431 cells are forced to express R-cadherin, they dramatically downregulate the expression of endogenous E- and P-cadherin. In addition, we show that this downregulation is owing to increased turnover of the endogenous cadherins via clathrin-dependent endocytosis. p120(ctn) binds to the juxtamembrane domain of classical cadherins and has been proposed to regulate cadherin adhesive activity. One way p120(ctn) may accomplish this is to serve as a rheostat to regulate the levels of cadherin. Here, we show that the degradation of E-cadherin in response to expression of R-cadherin is owing to competition for p120(ctn).
Collapse
Affiliation(s)
- M Maeda
- Department of Oral Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Yu JC, Hsu HM, Chen ST, Hsu GC, Huang CS, Hou MF, Fu YP, Cheng TC, Wu PE, Shen CY. Breast cancer risk associated with genotypic polymorphism of the genes involved in the estrogen-receptor-signaling pathway: a multigenic study on cancer susceptibility. J Biomed Sci 2006; 13:419-32. [PMID: 16502042 DOI: 10.1007/s11373-006-9069-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Accepted: 01/02/2006] [Indexed: 01/29/2023] Open
Abstract
The reproductive hormone, estrogen, contributes to the development of breast cancer by binding to the estrogen receptor (ER) in the nucleus, triggering cell growth and tumor promotion. In addition to its role in regulating target genes and signaling pathways involved in cell cycle progression, the ER-signaling pathway may regulate the expression of chromatin-remodeling gene, Metastasis-associated 3 (MTA3), or interact with chromatin-remodeling protein, Metastasis-associated 1 (MTA1). The invasion-suppressor gene, E-Cadherin (E-Cad), has recently been identified as a downstream target gene regulated by the ER-MTA3 pathway via the transcriptional repressor, Snail, and the ER-MTA3-Snail-E-Cad pathway has therefore been evoked to explain the clinical observation that ER expression in breast cancer is generally associated with a better clinical outcome. Since E-Cad may play an initiating role during breast tumorigenesis, we hypothesized that this ER-signaling pathway may also determine susceptibility to breast cancer, and examined this in a multigenic case-control study of 468 incident breast cancer patients and 470 healthy controls by genotyping the single nucleotide polymorphisms (SNPs) in five genes (ER, MTA3, Snail, E-Cad, and MTA1) in the ER-signaling pathways. Support for this hypothesis came from the observations that (a) with the exception of Snail, which interacted differently with reproductive risk factors in relation to breast cancer risk, there was a joint effect of the SNPs of these genes and estrogen-related risk factors (age at first full-term pregnancy and obesity, measured by the body mass index) on breast cancer risk (p < 0.05); (b) a trend toward increased risk of developing breast cancer was seen in women harboring a greater number of putative high-risk genotypes of these genes in ER-signaling pathways; (c) this association between risk and the number of putative high-risk genotypes was stronger and more significant in women thought to have experienced higher estrogen level, i.e., obese women; and (d) the risk effect conferred by obesity was only significant in women with a higher number of putative high-risk genotypes of the ER-signaling genes. These epidemiological findings highlight the role of newly identified novel ER-related pathways in breast cancer development and provide a more comprehensive picture of the tumorigenic effect of estrogen in breast cancer development.
Collapse
Affiliation(s)
- Jyh-Cherng Yu
- Departments of Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hung CF, Chiang HS, Lo HM, Jian JS, Wu WB. E-cadherin and its downstream catenins are proteolytically cleaved in human HaCaT keratinocytes exposed to UVB. Exp Dermatol 2006; 15:315-21. [PMID: 16512879 DOI: 10.1111/j.0906-6705.2006.00411.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It has been reported that ultraviolet B (UVB) irradiation causes the loss of E-cadherin of melanocytes, leading them to escape from neighboring keratinocytes during melanoma development. However, little has been paid on its effect on E-cadherin of keratinocytes. In the present study we therefore focus on whether UVB affects expression of E-cadherin-catenin complex in human HaCaT keratinocytes. We found that E-cadherin, beta-, and gamma-catenin but not alpha-catenin were proteolytically cleaved in UVB-irradiated HaCaT keratinocytes. The effect was only observed in keratinocyte undergoing apoptosis. Cleavage of beta- and gamma-catenin was fully abolished by caspase-3 and caspase-8 inhibitors, whereas cleavage of E-cadherin was inhibited by neither caspase nor metalloproteinase inhibitors. Functional analysis showed that the cleavage resulted in the disruption of the physical association between E-cadherin and catenins, indicating that E-cadherin signaling was compromised in UVB-irradiated HaCaT keratinocytes. Because E-cadherin in keratinocytes plays important roles in mediating cell-cell adhesion in epidermis of skin, the loss of E-cadherin and signaling components in keratinocytes may lead to the disruption of skin integrity after UVB exposure.
Collapse
Affiliation(s)
- Chi-Feng Hung
- School of Medicine, Fu-Jen Catholic University, Taipei County, Taiwan
| | | | | | | | | |
Collapse
|
39
|
Pećina-Slaus N, Kljaić M, Nikuseva-Martić T. Loss of heterozygosity of APC and CDH1 genes in laryngeal squamous cell carcinoma. Pathol Res Pract 2005; 201:557-63. [PMID: 16259108 DOI: 10.1016/j.prp.2005.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The molecular mechanisms involved in the development and progression of laryngeal cancer, specifically squamous cell carcinoma, still need further investigation and elucidation. Twenty-two laryngeal squamous cell carcinomas were analyzed in our study regarding genetic changes of two tumor suppressor genes: Adenomatous polyposis coli (APC) and E-cadherin (CDH1). APC gene instability was tested by polymerase chain reaction (PCR)/loss of heterozygosity (LOH) using the restriction fragment length polymorphism (RFLP) method. The samples were also screened for mutations using the heteroduplex method. E-cadherin gene was analyzed by PCR amplification of tetranucleotide marker (D16S752) linked to E-cadherin gene. The results of our analysis showed three samples with LOH of the APC gene out of 15 heterozygous patients (20%). Only one LOH of the CDH1 gene (5.5%) out of 18 heterozygous patients was discovered. D16S752 marker did not reveal any replication error-positive samples. There were six samples showing heteroduplexes (33%) encompassed in APC's exon 11. Altogether, nine samples (41%) showed alterations of the APC gene. Our results suggest that alterations of APC gene may have a role in squamous cell carcinoma development. Detected LOH of the E-cadherin gene indicates that genetic changes of this gene are not very frequent, but that other components of the wnt signaling cascade may also be involved.
Collapse
Affiliation(s)
- Nives Pećina-Slaus
- Department of Biology, School of Medicine, University of Zagreb, Croatia.
| | | | | |
Collapse
|
40
|
Romiti A, Zullo A, Borrini F, Sarcina I, Hassan C, Winn S, Tomao S, Vecchione A, Morini S, Mingazzini P. Relationship between β-catenin expression and epithelial cell proliferation in gastric mucosa with intestinal metaplasia. World J Gastroenterol 2005; 11:4400-3. [PMID: 16038041 PMCID: PMC4434669 DOI: 10.3748/wjg.v11.i28.4400] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate β-catenin expression in patients with intestinal metaplasia, and to look for a possible relationship between β-catenin expression and either epithelial proliferation values or Helicobacter pylori (H pylori) infection.
METHODS: Twenty patients with complete type intestinal metaplasia were studied. β-catenin expression and epithelial cell proliferation in antral mucosa were assessed using an immunohistochemical analysis. H pylori infection was detected by histology and a rapid urease test.
RESULTS: Reduced β-catenin expression on the surface of metaplastic cells was detected in 13 (65%) out of 20 patients. Moreover, in eight (40%) patients intranuclear expression of β-catenin was found. When patients were analyzed according to H pylori infection, the prevalence of both β-catenin reduction at the cell surface and its intranuclear localization did not significantly differ between infected and uninfected patients. Cell proliferation was higher in patients with intranuclear β-catenin expression as compared to the remaining patients, although the difference failed to reach the statistical significance (36 ± 8.9 vs 27.2 ± 11.4, P = 0.06). On the contrary, a similar cell proliferation value was observed between patients with reduced expression of β-catenin on cell surface and those with a normal expression (28.1 ± 11.8 vs 26.1 ± 8.8, P = 0.7). H pylori infection significantly increased cell proliferation (33.3 ± 10.2% vs 24.6 ± 7.4%, respectively, P = 0.04).
CONCLUSION: Both cell surface reduction and intranuclear accumulation of β-catenin were detected in intestinal metaplasia. The intranuclear localization of β-catenin increases cell proliferation. H pylori infection does not seem to play a direct role in β-catenin alterations, whilst it significantly increases cell proliferation.
Collapse
|
41
|
Shin SJ, Kim KO, Kim MK, Lee KH, Hyun MS, Kim KJ, Choi JH, Song HS. Expression of E-Cadherin and uPA and their Association with the Prognosis of Pancreatic Cancer. Jpn J Clin Oncol 2005; 35:342-8. [PMID: 15937032 DOI: 10.1093/jjco/hyi094] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE E-cadherin (ECD) and urokinase plasminogen activator (uPA) have been noted as markers for tumor metastasis and prognosis in several tumors. We thus investigated the relationship between the expression of ECD and uPA and the clinicopathological characteristics in pancreatic cancer. METHODS The expression of ECD and uPA was evaluated in pancreatic cancer tissues from 53 patients. RESULTS Among 53 tumor tissues, those from 29 (54.7%) patients showed positive ECD expression and those from 22 (41.5%) patients showed positive expression of uPA. There were four subgroups of ECD/uPA expression: ECD-positive/uPA-negative, ECD-negative/uPA-negative, ECD-positive/uPA-positive and ECD-negative/uPA-positive. These patterns were found in 14 (26.4%), 11 (20.8%), nine (17%) and 19 (35.8%) patients, respectively. The tumor tissues with ECD-negative and uPA-positive expression were associated with larger tumor, distant metastasis and an increased clinical stage. There was a difference in the median survival time between the patients with ECD-positive/uPA-negative pancreatic tissues (median: 18.7 months) and the patients with ECD-negative/uPA-positive pancreatic tissues (median: 7.5 months, P < 0.05), and there was a statistically significant difference in survival curves between these two groups. CONCLUSION The combined analysis concerning uPA and E-cadherin expression may be a useful predictor of metastasis in pancreatic cancer.
Collapse
Affiliation(s)
- Sang Joon Shin
- Division of Oncology-Hematology, Department of Medicine, Yeungnam University College of Medicine, Daemyeung-Dong, 317-1, Namg-Gu, Daegu 705-717, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Eriksen JG, Steiniche T, Overgaard J. The role of epidermal growth factor receptor and E-cadherin for the outcome of reduction in the overall treatment time of radiotherapy of supraglottic larynx squamous cell carcinoma. Acta Oncol 2005; 44:50-8. [PMID: 15848906 DOI: 10.1080/02841860510007396] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Reduction of the overall treatment time (OTT) of radiotherapy results in increased T-site control in squamous cell carcinomas of the head and neck (HNSCC). However, the response is heterogeneous and accelerated repopulation of clonogenic tumour cells during therapy may be one of the factors determining this response. The aim of the present study was to identify the influence of the epidermal growth factor receptor (EGFr) and E-cadherin for T-site control when the OTT was reduced and whether the markers add information to the histopathological grading in selecting patients for accelerated radiotherapy. A total of 209 patients from randomized DAHANCA-trials with supraglottic larynx squamous cell carcinomas treated with primary radiotherapy with different OTT of 9(1/2), 6(1/2), and 5(1/2) weeks. Available formalin-fixed paraffin embedded tumour tissues were re-evaluated for histopathological characteristics and stained for EGFr and E-cadherin. Data were correlated with patient and tumour characteristics and 5-year T-site control. EGFr and E-cadherin were not associated with patient or tumour characteristics except that EGFr correlated to carcinomas with a well to moderate histopathological feature. Tumours with high EGFr or low E-cadherin did benefit from reduced OTT, and the combination of the two (high EGFr and low E-cadherin) had the most significant acceleration of treatment effect, compared with tumours with other combinations of EGFr and E-cadherin expression. Tumours with high expression of EGFr and low expression of E-cadherin showed the most significant increase in T-site control when the overall treatment time of radiotherapy was reduced, and the markers may be useful for selecting patients who will benefit from accelerated radiotherapy.
Collapse
Affiliation(s)
- Jesper Grau Eriksen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark.
| | | | | |
Collapse
|
43
|
Moran CJ, Joyce M, McAnena OJ. CDH1 associated gastric cancer: a report of a family and review of the literature. Eur J Surg Oncol 2005; 31:259-64. [PMID: 15780560 DOI: 10.1016/j.ejso.2004.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2004] [Indexed: 12/11/2022] Open
Abstract
AIMS Hereditary diffuse gastric cancer (HDGC) is an autosomal-dominant inherited form of gastric cancer associated with inactivating germline mutations in the CDH1 gene. We set out to outline the role of CDH1 in HDGC. Investigation of a family suspected as having HDGC is discussed. The role of surgery in the management of affected individuals is then examined. METHODS A search was conducted of Medline and the National Library of Medicine to identify key articles concerning CDH1 gene mutations, familial gastric cancer and gastrectomy. Further, relevant articles were obtained by manual scanning of the reference lists of identified papers. Mutation-specific CDH1 genetic testing was performed on six living family members and on gastric tissue obtained from two deceased members. RESULTS CDH1 mutations cause inactivation of the cell adhesion protein E-cadherin. Carriers of the CDH1 germline gene mutation develop an aggressive, diffuse, submucosal gastric cancer at an early age. Current endoscopic screening is ineffective at detecting HDGC. The presence of a CDH1 germline gene mutation was confirmed in both deceased family members and also in four of the six living members tested. CONCLUSION Genetic counselling and CDH1 gene mutation testing is indicated in families with suspected HDGC. In the absence of a satisfactory surveillance mechanism, prophylactic total gastrectomy would appear to be an appropriate therapeutic option in mutation carriers.
Collapse
Affiliation(s)
- C J Moran
- Department of Surgery, University College Hospital Galway, Newcastle Road, Galway, Ireland.
| | | | | |
Collapse
|
44
|
Lee DW, Park SW, Park SY, Heo DS, Kim KH, Sung MW. Effects of p53 or p27 overexpression on cyclooxygenase-2 gene expression in head and neck squamous cell carcinoma cell lines. Head Neck 2005; 26:706-15. [PMID: 15287038 DOI: 10.1002/hed.20025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Although cyclooxygenase-2 (COX-2) has been suggested to play an important role in carcinogenesis, the effects of tumor suppressors on COX-2 gene expression and the combined antitumor effects of tumor suppressors and COX-2 inhibitors have rarely been investigated. METHODS The effects of p53 or p27 gene transfer on COX-2 expression by adenoviral vector and the combined effects of p53 or p27 gene transfer and COX-2 inhibitor exposure on the proliferation of cancer cells were investigated in head and neck squamous cell carcinoma (HNSCC) cell lines. RESULTS Overexpression of p53 markedly downregulated the transcription of COX-2, but the overexpression of p27 did not affect COX-2 levels in HNSCC cell lines. The combined antitumor effects of p53 or p27 gene transfer and of a COX-2 inhibitor (NS 398) were mainly at least additive in terms of the inhibition of cell proliferation and cell cycle arrest and additive in terms of apoptotic induction. CONCLUSIONS These results suggest that the overexpression of p53 could exert antitumor effects, at least in part, through the suppression of COX-2 gene expression, whereas growth suppression by the overexpression of p27 probably occurs by mechanisms other than the downregulation of COX-2 expression. In addition, the administration of COX-2 inhibitors, as an adjunct to p53 or p27 gene therapy, could offer a new strategy of cancer treatment and prevention.
Collapse
MESH Headings
- Analysis of Variance
- Apoptosis
- Blotting, Western
- Carcinoma, Squamous Cell/enzymology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Cycle/physiology
- Cell Cycle Proteins/genetics
- Cell Line, Tumor
- Cell Proliferation
- Cyclin-Dependent Kinase Inhibitor p27
- Cyclooxygenase 2
- Cyclooxygenase 2 Inhibitors
- Cyclooxygenase Inhibitors
- Down-Regulation
- Gene Expression Regulation, Enzymologic/genetics
- Gene Expression Regulation, Neoplastic/genetics
- Genes, Tumor Suppressor/physiology
- Genes, p53/physiology
- Head and Neck Neoplasms/enzymology
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/pathology
- Humans
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Membrane Proteins
- Promoter Regions, Genetic/genetics
- Prostaglandin-Endoperoxide Synthases/genetics
- Prostaglandin-Endoperoxide Synthases/metabolism
- Transfection
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- Dong Wook Lee
- Department of Otorhinolaryngology-Head and Neck Surgery and Medical Research Institute, College of Medicine, Chungbuk National University, Cheongju, Korea
| | | | | | | | | | | |
Collapse
|
45
|
Martinez-Rico C, Pincet F, Perez E, Thiery JP, Shimizu K, Takai Y, Dufour S. Separation Force Measurements Reveal Different Types of Modulation of E-cadherin-based Adhesion by Nectin-1 and -3. J Biol Chem 2005; 280:4753-60. [PMID: 15550395 DOI: 10.1074/jbc.m412544200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nectins are Ca2+-independent cell adhesion molecules found at cadherin-based adherens junctions. We used a dual pipette assay that measures the forces required to separate cell doublets to determine how nectins affect the formation and strength of cell-cell adhesion. Less force was required to separate doublets of L cells expressing nectin-1 or nectin-3 than to separate doublets of E-cadherin-expressing cells. Heterodimers formed between cells expressing nectin-1 or nectin-3 adhered more strongly than homodimers. Nectin-3 that does not trans-interact with nectin-1 inhibited E-cadherin-mediated adhesion. However, the extracellular fragment of nectin-1 did not have an agonistic effect on E-cadherin-dependent cell adhesion when it trans-interacted with nectin-3, expressed at high levels in cells. In contrast, the extracellular fragment of nectin-3 had a significant agonistic effect on cadherin-based adhesion when it interacted with endogenous nectin-1, expressed at low levels in cells. Our results indicate that E-cadherin is the key molecule involved in cell adhesion and that the regulation of E-cadherin-based adhesion involving cellular nectin-1 trans-interacting with nectin-3 is qualitatively different from that involving cellular nectin-3 trans-interacting with nectin-1 and depends on the nectin levels expressed by cells.
Collapse
|
46
|
Abstract
Under normal homeostasis, melanocyte growth and behaviour is tightly controlled by the surrounding keratinocytes. Keratinocytes regulate melanocyte behaviour through a complex system of paracrine growth factors and cell-cell adhesion molecules. Pathological changes, leading to development of malignant melanoma, upset this delicate homeostatic balance and can lead to altered expression of cell-cell adhesion and cell-cell communication molecules. In particular, there is a switch from the E-cadherin-mediated keratinocyte-melanocyte partnership to the N-cadherin-mediated melanoma-melanoma and melanoma-fibroblast interaction. Other changes include the alteration in the gap junctions formed between the melanocyte and keratinocyte. Changes in the connexin expression, in particular the loss of connexin 43, may result in a reduction or a loss of gap junctional activity, which is thought to contribute towards tumour progression. In the current review we describe the alterations in cell-cell adhesion and communication associated with melanoma development and progression, and discuss how a greater understanding of these processes may aid the future therapy of this disease.
Collapse
Affiliation(s)
- Nikolas K Haass
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
47
|
Graziano F, Arduini F, Ruzzo A, Bearzi I, Humar B, More H, Silva R, Muretto P, Guilford P, Testa E, Mari D, Magnani M, Cascinu S. Prognostic analysis of E-cadherin gene promoter hypermethylation in patients with surgically resected, node-positive, diffuse gastric cancer. Clin Cancer Res 2004; 10:2784-9. [PMID: 15102685 DOI: 10.1158/1078-0432.ccr-03-0320] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Recent investigations have demonstrated that hypermethylation is a frequent mechanism for silencing tumor suppressor genes. This is a potentially reversible epigenetic change, and it is the target of a novel class of anticancer compounds with demethylating activity. Better understanding of the clinical implications of hypermethylation will allow the optimal planning of future trials with demethylating drugs. In this perspective, we investigated whether hypermethylation in the CDH1 promoter region is correlated with poor prognosis of patients with surgically resected, node-positive, diffuse gastric cancer. EXPERIMENTAL DESIGN Consecutive cases of diffuse gastric cancer were considered eligible for study entry. Additional inclusion criteria were radical surgery with a minimum of D1 lymphadenectomy, complete follow-up information, and availability of tumor specimens for methylation-specific PCR and immunohistochemistry analyses. RESULTS CDH1 promoter hypermethylation was found in 40 of 73 cases (54%), and it was significantly associated with worse prognosis. In patients with and without hypermethylation, the 5-year event-free survival rate was 30% and 62%, respectively, and the 5-year overall survival rate was 35% and 67%, respectively. CDH1 promoter hypermethylation retained its prognostic role for disease-free survival (P < 0.001) and overall survival (P < 0.001) in multivariate analysis. Immunohistochemistry showed a significant association between CDH1 methylation and E-cadherin expression (P < 0.001). CONCLUSIONS This study shows adverse prognostic effect of CDH1 promoter hypermethylation in patients with diffuse gastric cancer. This form of cancer, and other types with frequent hypermethylation and silencing of critical tumor suppressor genes, would make appropriate targets for the testing of novel compounds with demethylating activity.
Collapse
|
48
|
Eriksen JG, Steiniche T, Søgaard H, Overgaard J. Expression of integrins and E-cadherin in squamous cell carcinomas of the head and neck. APMIS 2004; 112:560-8. [PMID: 15601304 DOI: 10.1111/j.1600-0463.2004.apm1120902.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Integrins and cadherins are cell adhesion molecules suggested to play an important role in malignant progression and tumour differentiation. Our aim was to characterise the pattern of expression and the relations between integrin beta1, beta4, beta6 and E-cadherin and the different histopathological features important when judging tumour differentiation, using a well-defined scoring system. Formalin-fixed paraffin-embedded pre-irradiation biopsies from 85 patients with head and neck squamous cell carcinomas (HNSCC) were stained and evaluated for the expression of integrin beta1, beta4 and beta6 and E-cadherin. The integrins were upregulated in carcinomas compared to the adjacent mucosa and E-cadherin was downregulated. However, differences were found within the tumour: Expression of E-cadherin was lost and the three integrins were upregulated at the tumour borders, compared to central parts of the tumour biopsy. Expression of the integrins did not correlate with tumour or histopathological parameters, whereas expression of E-cadherin was correlated with high degree of keratinisation, high nuclear maturation and few mitoses - factors that characterise well-differentiated carcinomas -and E-cadherin can therefore be considered as a marker of differentiation. Furthermore, loss of adhesion expressed by low E-cadherin and integrin beta4 correlated with the presence of nodal metastases at the time of diagnosis.
Collapse
Affiliation(s)
- J G Eriksen
- Department of Experimental Clinical Oncology, University Hospital of Aarhus, Denmark.
| | | | | | | |
Collapse
|
49
|
Pijnenborg JMA, Kisters N, van Engeland M, Dunselman GAJ, de Haan J, de Goeij AFPM, Groothuis PG. APC, beta-catenin, and E-cadherin and the development of recurrent endometrial carcinoma. Int J Gynecol Cancer 2004; 14:947-56. [PMID: 15361208 DOI: 10.1111/j.1048-891x.2004.014534.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Endometrial carcinoma, generally, has a good prognosis. However, in some patients, the tumor appears to behave very aggressively, a course that cannot be explained with histopathological characteristics. More insight into the molecular background can be valuable to clarify these differences in tumor behavior. The three components associated with the Wnt pathway--i.e., adenomatous polyposis coli (APC), beta-catenin, and E-cadherin--were evaluated in a case-control study of 28 patients with stage-I endometrial carcinomas to determine their involvement in the development of recurrent disease. Mutation analysis of the mutation cluster region of the APC gene, determination of gene promoter methylation status of the APC-1A and E-cadherin genes, and immunohistochemical analysis of APC, E-cadherin, and beta-catenin were performed using paraffin-embedded tumor tissue. Twenty-one APC gene mutations were detected in 12 of 28 (43%) patients. Only three mutations would result in a stopcodon in the APC gene. APC gene promoter methylation was assessed in 12 of 28 (43%) patients. APC immunostaining was absent in two of 24 (8.3%) patients. The occurrence of APC mutations, APC gene promoter methylation, and APC immunostaining were not predictive for recurrence. No E-cadherin expression was observed in four of 24 patients (17%). E-cadherin gene promoter methylation could not be detected in any of the patients. The absence of E-cadherin expression was predictive for distant metastases, but not for local recurrence. Nuclear localization of beta-catenin was present in nine of 24 (38%) patients and was not predictive for recurrent disease. Involvement of epigenetic and genetic aberrations in APC and beta-catenin genes seems to be of minor importance for the development of local recurrences and distant metastases. Although the number of patients is limited, E-cadherin expression appears to be predictive for the development of distant metastases in endometrial carcinoma.
Collapse
Affiliation(s)
- J M A Pijnenborg
- Department of Obstetrics and Gynecology, University Hospital Maastricht and University of Maastricht, Maastricht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
50
|
Li G, Fukunaga M, Herlyn M. Reversal of melanocytic malignancy by keratinocytes is an E-cadherin-mediated process overriding beta-catenin signaling. Exp Cell Res 2004; 297:142-51. [PMID: 15194432 DOI: 10.1016/j.yexcr.2004.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2003] [Revised: 02/02/2004] [Indexed: 01/01/2023]
Abstract
Loss of E-cadherin in melanoma cells frees them from keratinocytes-mediated proliferation and phenotypic control, which can be restored by forced E-cadherin expression. In this study, E-cadherin and its derivatives were introduced into metastatic melanoma line 1205Lu. E-cadherin and E-cadherin-alpha-catenin fusion protein were functional in mediating cell adhesion, downregulating MCAM(4) in coculture, and inhibiting proliferation regardless of beta-catenin expression levels and activation status. In contrast, cytoplasmic domain-deleted (E-cadDeltaCYT) derivative was not able to reverse malignancy. The results indicate that E-cadherin-mediated cell adhesion is required for keratinocyte-mediated control of melanocytic cells, which can override proliferative activity of beta-catenin.
Collapse
Affiliation(s)
- Gang Li
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|