1
|
Arafeh R, Shibue T, Dempster JM, Hahn WC, Vazquez F. The present and future of the Cancer Dependency Map. Nat Rev Cancer 2025; 25:59-73. [PMID: 39468210 DOI: 10.1038/s41568-024-00763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/30/2024]
Abstract
Despite tremendous progress in the past decade, the complex and heterogeneous nature of cancer complicates efforts to identify new therapies and therapeutic combinations that achieve durable responses in most patients. Further advances in cancer therapy will rely, in part, on the development of targeted therapeutics matched with the genetic and molecular characteristics of cancer. The Cancer Dependency Map (DepMap) is a large-scale data repository and research platform, aiming to systematically reveal the landscape of cancer vulnerabilities in thousands of genetically and molecularly annotated cancer models. DepMap is used routinely by cancer researchers and translational scientists and has facilitated the identification of several novel and selective therapeutic strategies for multiple cancer types that are being tested in the clinic. However, it is also clear that the current version of DepMap is not yet comprehensive. In this Perspective, we review (1) the impact and current uses of DepMap, (2) the opportunities to enhance DepMap to overcome its current limitations, and (3) the ongoing efforts to further improve and expand DepMap.
Collapse
Affiliation(s)
- Rand Arafeh
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | | | | | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
2
|
Moya L, Walpole C, Rae F, Srinivasan S, Seim I, Lai J, Nicol D, Williams ED, Clements JA, Batra J. Characterisation of cell lines derived from prostate cancer patients with localised disease. Prostate Cancer Prostatic Dis 2023; 26:614-624. [PMID: 37264224 PMCID: PMC10449630 DOI: 10.1038/s41391-023-00679-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 04/17/2023] [Accepted: 05/12/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Prostate cancer is a broad-spectrum disease, spanning from indolent to a highly aggressive lethal malignancy. Prostate cancer cell lines are essential tools to understanding the basic features of this malignancy, as well as in identifying novel therapeutic strategies. However, most cell lines routinely used in prostate cancer research are derived from metastatic disease and may not fully elucidate the molecular events underlying the early stages of cancer development and progression. Thus, there is a need for new cell lines derived from localised disease to better span the disease spectrum. METHODS Prostatic tissue from the primary site, and adjacent non-cancerous tissue was obtained from four patients with localised disease undergoing radical prostatectomy. Epithelial cell outgrowths were immortalised with human papillomavirus type 16 (HPV16) E6 and E7 to establish monoclonal cell lines. Chromosomal ploidy was imaged and STR profiles were determined. Cell morphology, colony formation and cell proliferation characteristics were assessed. Androgen receptor (AR) expression and AR-responsiveness to androgen treatment were analysed by immunofluorescence and RT-qPCR, respectively. RNA-seq analysis was performed to identify prostate lineage markers and expression of prostate cancer tumorigenesis-related genes. RESULTS Two benign cell lines derived from non-cancer cells (AQ0420 and AQ0396) and two tumour tissue derived cancer cell lines (AQ0411 and AQ0415) were immortalised from four patients with localised prostatic adenocarcinoma. The cell lines presented an epithelial morphology and a slow to moderate proliferative rate. None of the cell lines formed anchorage independent colonies or displayed AR-responsiveness. Comparative RNA-seq expression analysis confirmed the prostatic lineage of the four cell lines, with a distinct gene expression profile from that of the metastatic prostate cancer cell lines, PC-3 and LNCaP. CONCLUSIONS Comprehensive characterization of these cell lines may provide new in vitro tools that could bridge the current knowledge gap between benign, early-stage and metastatic disease.
Collapse
Affiliation(s)
- Leire Moya
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Carina Walpole
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
- Cancer Immunotherapies Group, Mater Research, Translational Research Institute, Brisbane, Australia
| | - Fiona Rae
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Inge Seim
- Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing, China
- School of Biology and Environmental Science, Queensland University of Technology, Brisbane, Australia
| | - John Lai
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
- Australian Genome Research Facility Ltd, Gehrmann Laboratories, the University of Queensland, Brisbane, Australia
| | - David Nicol
- Urology Department, Princess Alexandra Hospital, Brisbane, Australia
- Urology Unit, The Royal Marsden, London, UK
| | - Elizabeth D Williams
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
- Department of Surgery, St Vincent's Hospital, University of Melbourne, Melbourne, Australia
| | - Judith A Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.
- Center for genomics and Personalised Health, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
3
|
Sailer V, von Amsberg G, Duensing S, Kirfel J, Lieb V, Metzger E, Offermann A, Pantel K, Schuele R, Taubert H, Wach S, Perner S, Werner S, Aigner A. Experimental in vitro, ex vivo and in vivo models in prostate cancer research. Nat Rev Urol 2023; 20:158-178. [PMID: 36451039 DOI: 10.1038/s41585-022-00677-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 12/02/2022]
Abstract
Androgen deprivation therapy has a central role in the treatment of advanced prostate cancer, often causing initial tumour remission before increasing independence from signal transduction mechanisms of the androgen receptor and then eventual disease progression. Novel treatment approaches are urgently needed, but only a fraction of promising drug candidates from the laboratory will eventually reach clinical approval, highlighting the demand for critical assessment of current preclinical models. Such models include standard, genetically modified and patient-derived cell lines, spheroid and organoid culture models, scaffold and hydrogel cultures, tissue slices, tumour xenograft models, patient-derived xenograft and circulating tumour cell eXplant models as well as transgenic and knockout mouse models. These models need to account for inter-patient and intra-patient heterogeneity, the acquisition of primary or secondary resistance, the interaction of tumour cells with their microenvironment, which make crucial contributions to tumour progression and resistance, as well as the effects of the 3D tissue network on drug penetration, bioavailability and efficacy.
Collapse
Affiliation(s)
- Verena Sailer
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Gunhild von Amsberg
- Department of Oncology and Hematology, University Cancer Center Hamburg Eppendorf and Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Stefan Duensing
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg and National Center for Tumour Diseases, Heidelberg, Germany
| | - Jutta Kirfel
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Verena Lieb
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Eric Metzger
- Department of Urology, Center for Clinical Research, University of Freiburg Medical Center, Freiburg, Germany
| | - Anne Offermann
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Klaus Pantel
- Institute for Tumour Biology, Center for Experimental Medicine, University Clinics Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel-Nachwuchszentrum HaTRiCs4, University Cancer Center Hamburg, Hamburg, Germany
| | - Roland Schuele
- Department of Urology, Center for Clinical Research, University of Freiburg Medical Center, Freiburg, Germany
| | - Helge Taubert
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Sven Wach
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Sven Perner
- University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Stefan Werner
- Institute for Tumour Biology, Center for Experimental Medicine, University Clinics Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel-Nachwuchszentrum HaTRiCs4, University Cancer Center Hamburg, Hamburg, Germany
| | - Achim Aigner
- Clinical Pharmacology, Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Medical Faculty, Leipzig, Germany.
| |
Collapse
|
4
|
Hong JH, Liang ST, Wang ASS, Yeh CM, Huang HP, Sun CD, Zhang ZH, Lu SY, Chao YH, Chen CH, Pu YS. LMNB1, a potential marker for early prostate cancer progression. Am J Cancer Res 2022; 12:3390-3404. [PMID: 35968338 PMCID: PMC9360214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/20/2022] [Indexed: 06/15/2023] Open
Abstract
Although prostate cancer (PC) is the most common cancer among men in the Western world, there are no good biomarkers that can reliably differentiate between potentially aggressive and indolent PC. This leads to overtreatment, even for patients who can be managed conservatively. Previous studies have suggested that nuclear lamin proteins-especially lamin B1 (LMNB1)-play important roles in PC progression. However, the results of these studies are inconsistent. Here, we transfected the LMNB1 gene into the telomerase reverse transcriptase-immortalized benign prostatic epithelial cell line, EP156T to generate a LMNB1-overexpressing EP156T (LMN-EP156T) cell line with increased cellular proliferation. However, LMN-EP156T cells could neither form colonies in soft agar, nor establish subcutaneous growth or metastasis in the xenograft NOD/SCID mouse model. In addition, immunohistochemical staining of LMNB1 in PC specimens from 143 patients showed a statistically significant trend of stronger LMNB1 staining with higher Gleason scores. A univariate analysis of the clinicopathological parameters of 85 patients with PC who underwent radical prostatectomy revealed that pathological stage, resection margin, and extracapsular extension were significant predictors for biochemical recurrence (BCR). However, LMNB1 staining showed only a non-significant trend of association with BCR (high vs. low staining: hazard ratio (HR), 1.83; 95% confidence interval (CI), 0.98-3.41; P = 0.059). In multivariate analysis, only pathological stage was a significant independent predictor of BCR (pT3 vs. pT2: HR, 2.29; 95% CI, 1.18-4.43; P = 0.014). In summary, LMNB1 may play a role in the early steps of PC progression, and additional molecular alterations may be needed to confer full malignancy potential to initiated cells.
Collapse
Affiliation(s)
- Jian-Hua Hong
- Department of Urology, National Taiwan University HospitalTaipei, Taiwan
- Institute of Biomedical Engineering, National Taiwan UniversityTaipei, Taiwan
| | - Sung-Tzu Liang
- Department of Urology, National Taiwan University HospitalTaipei, Taiwan
| | | | - Chia-Ming Yeh
- Department of Urology, National Taiwan University HospitalTaipei, Taiwan
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of MedicineTaipei, Taiwan
| | - Chia-Dong Sun
- Department of Pathology, National Taiwan University HospitalTaipei, Taiwan
- Department and Graduate Institute of Forensic Medicine, College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | | | - Shih-Yu Lu
- Department of Urology, National Taiwan University HospitalTaipei, Taiwan
| | - Yen-Hsiang Chao
- Department of Urology, National Taiwan University HospitalTaipei, Taiwan
| | - Chung-Hsin Chen
- Department of Urology, National Taiwan University HospitalTaipei, Taiwan
| | - Yeong-Shiau Pu
- Department of Urology, National Taiwan University HospitalTaipei, Taiwan
| |
Collapse
|
5
|
Azeem W, Olsen JR, Hellem MR, Hua Y, Marvyin K, Ke X, Øyan AM, Kalland KH. Proteasome-Mediated Regulation of GATA2 Expression and Androgen Receptor Transcription in Benign Prostate Epithelial Cells. Biomedicines 2022; 10:biomedicines10020473. [PMID: 35203681 PMCID: PMC8962351 DOI: 10.3390/biomedicines10020473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
GATA2 has been shown to be an important transcription factor together with androgen receptor (AR) in prostate cancer cells. Less is known about GATA2 in benign prostate epithelial cells. We have investigated if GATA2 exogenous expression in prostate epithelial basal-like cells could induce AR transcription or luminal differentiation. Prostate epithelial basal-like (transit amplifying) cells were transduced with lentiviral vector expressing GATA2. Luminal differentiation markers were assessed by RT-qPCR, Western blot and global gene expression microarrays. We utilized our previously established AR and androgen-dependent fluorescence reporter assay to investigate AR activity at the single-cell level. Exogenous GATA2 protein was rapidly and proteasome-dependently degraded. GATA2 protein expression was rescued by the proteasome inhibitor MG132 and partly by mutating the target site of the E3 ligase FBXW7. Moreover, MG132-mediated proteasome inhibition induced AR mRNA and additional luminal marker gene transcription in the prostate transit amplifying cells. Different types of intrinsic mechanisms restricted GATA2 expression in the transit amplifying cells. The appearance of AR mRNA and additional luminal marker gene expression changes following proteasome inhibition suggests control of essential cofactor(s) of AR mRNA expression and luminal differentiation at this proteolytic level.
Collapse
Affiliation(s)
- Waqas Azeem
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence: (W.A.); (K.-H.K.)
| | - Jan Roger Olsen
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Margrete Reime Hellem
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Yaping Hua
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Kristo Marvyin
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Xisong Ke
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Anne Margrete Øyan
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Karl-Henning Kalland
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence: (W.A.); (K.-H.K.)
| |
Collapse
|
6
|
Zhao Z, Fowle H, Valentine H, Liu Z, Tan Y, Pei J, Badal S, Testa JR, Graña X. Immortalization of human primary prostate epithelial cells via CRISPR inactivation of the CDKN2A locus and expression of telomerase. Prostate Cancer Prostatic Dis 2021; 24:233-243. [PMID: 32873916 PMCID: PMC7917161 DOI: 10.1038/s41391-020-00274-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/10/2020] [Accepted: 08/19/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Immortalization of primary prostate epithelial cells (PrEC) with just hTERT expression is particularly inefficient in the absence of DNA tumor viral proteins or p16INK4A knockdown. MATERIALS AND METHODS Here, we describe the establishment of immortalized normal prostate epithelial cell line models using CRISPR technology to inactivate the CDKN2A locus concomitantly with ectopic expression of an hTERT transgene. RESULTS Using this approach, we have obtained immortal cell clones that exhibit fundamental characteristics of normal cells, including diploid genomes, near normal karyotypes, normal p53 and pRB cell responses, the ability to form non-invasive spheroids, and a non-transformed phenotype. Based on marker expression, these clones are of basal cell origin. CONCLUSIONS Use of this approach resulted in the immortalization of independent clones of PrEC that retained normal characteristics, were stable, and non-transformed. Thus, this approach could be used for the immortalization of normal primary prostate cells. This technique could also be useful for establishing cell lines from prostate tumor tissues of different tumor grades and/or from patients of diverse ethnicities to generate cell line models that facilitate the study of the molecular basis of disease disparity.
Collapse
Affiliation(s)
- Ziran Zhao
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA
| | - Holly Fowle
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA
| | - Henkel Valentine
- Department of Basic Medical Sciences, Faculty of Medical Sciences Teaching and Research Complex, The University of the West Indies, Mona, Jamaica
| | - Zemin Liu
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Yinfei Tan
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Jianming Pei
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Simone Badal
- Department of Basic Medical Sciences, Faculty of Medical Sciences Teaching and Research Complex, The University of the West Indies, Mona, Jamaica
| | - Joseph R Testa
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Xavier Graña
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Ando T, Kage H, Matsumoto Y, Zokumasu K, Yotsumoto T, Maemura K, Amano Y, Watanabe K, Nakajima J, Nagase T, Takai D. Integrin α11 in non-small cell lung cancer is associated with tumor progression and postoperative recurrence. Cancer Sci 2019; 111:200-208. [PMID: 31778288 PMCID: PMC6942423 DOI: 10.1111/cas.14257] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/16/2019] [Accepted: 11/22/2019] [Indexed: 12/24/2022] Open
Abstract
Integrins are transmembrane proteins that mediate cell adhesion to the extracellular matrix. Integrin α11 (ITGA11) is not expressed in normal alveolar epithelial cells and is a known receptor for collagen. While integrin α11β1 overexpression in the tumor stroma has been associated with tumor growth and metastatic potential of non-small cell lung cancer (NSCLC), little is known about the role of ITGA11 in tumor cells. Thus, we examined the RNA expression of ITGA11 by quantitative RT-PCR in 80 samples collected from NSCLC patients who had undergone surgical resection and analyzed the clinical outcomes. We found that high expression of ITGA11 was associated with lower recurrence-free survival in all NSCLC patients (P = 0.043) and in stage I NSCLC patients (P = 0.049). These results were consistent with in silico analyses of the Cancer Genome Atlas database. We also analyzed cell proliferation, migration and invasion capacity in lung cancer cell lines after overexpression of ITGA11. Overexpression of ITGA11 in lung cancer cell lines had little effect on cell proliferation but resulted in increased migration and invasion capacity. Our findings suggest that ITGA11 plays a significant role in cancer migration and invasion, leading to higher recurrence. ITGA11 expression may be a predictor of poor prognosis in patients with surgically resected NSCLC.
Collapse
Affiliation(s)
- Takahiro Ando
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Hidenori Kage
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoko Matsumoto
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Koichi Zokumasu
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Takuma Yotsumoto
- Department of Thoracic Surgery, The University of Tokyo, Tokyo, Japan
| | - Keita Maemura
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Amano
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Kousuke Watanabe
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Nakajima
- Department of Thoracic Surgery, The University of Tokyo, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Daiya Takai
- Department of Clinical Laboratory, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Senchukova M, Tomchuk O, Shurygina E, Letuta S, Alidzhanov E, Nikiyan H, Razdobreev D. Calcium Carbonate Nanoparticles Can Activate the Epithelial⁻Mesenchymal Transition in an Experimental Gastric Cancer Model. Biomedicines 2019; 7:21. [PMID: 30893803 PMCID: PMC6466388 DOI: 10.3390/biomedicines7010021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 02/05/2023] Open
Abstract
Previously, we have shown the possibility of intramucosal gastric carcinoma induction by the intragastric administration of a mixture of formaldehyde and hydrogen peroxide in rats. In this study, we report a sizable increase in carcinogenic properties of the mixture when a suspension containing calcium carbonate nanoparticles was added to it. This technique allowed us to reduce both the number of the carcinogen administrations from twelve to two and the time to the cancer induction from six to four months. Although the induced tumors were represented by the intramucosal carcinomas, they were characterized by the extensive invasion of individual tumor cells and their clusters into the muscle layer and serosa as well as into the omentum and blood vessels. Considering that the invasive tumor cells were positive for vimentin, Snail and TGF-β2, we concluded that their invasion was the result of the activation of epithelial⁻mesenchymal transition (EMT) mechanisms. Thus, taking into account the data obtained, it can be assumed that under the conditions of inflammation or carcinogenesis, the calcium carbonate nanoparticles may affect the activation of EMT mechanisms.
Collapse
Affiliation(s)
- Marina Senchukova
- Department of Oncology, Orenburg State Medical University, Orenburg 460000, Russia.
| | - Olesya Tomchuk
- Department of Histology, Cytology, Embryology, Orenburg State Medical University, Orenburg 460000, Russia.
| | - Elena Shurygina
- Department of Pathology, Orenburg State Medical University, Orenburg 460000, Russia.
| | - Sergey Letuta
- Department of Biophysics and Condensed Matter Physics, Orenburg State University, Orenburg 460018, Russia.
- Institute of micro- and nanotechnology, Orenburg State University, Orenburg 460018, Russia.
| | - Eskender Alidzhanov
- Department of Biophysics and Condensed Matter Physics, Orenburg State University, Orenburg 460018, Russia.
- Institute of micro- and nanotechnology, Orenburg State University, Orenburg 460018, Russia.
| | - Hike Nikiyan
- Department of Biophysics and Condensed Matter Physics, Orenburg State University, Orenburg 460018, Russia.
- Institute of micro- and nanotechnology, Orenburg State University, Orenburg 460018, Russia.
| | - Dmitry Razdobreev
- Department of Biophysics and Condensed Matter Physics, Orenburg State University, Orenburg 460018, Russia.
- Institute of micro- and nanotechnology, Orenburg State University, Orenburg 460018, Russia.
| |
Collapse
|
9
|
Jones KM, Karanam B, Jones-Triche J, Sandey M, Henderson HJ, Samant RS, Temesgen S, Yates C, Bedi D. Phage Ligands for Identification of Mesenchymal-Like Breast Cancer Cells and Cancer-Associated Fibroblasts. Front Oncol 2019; 8:625. [PMID: 30619759 PMCID: PMC6304394 DOI: 10.3389/fonc.2018.00625] [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: 07/08/2018] [Accepted: 12/03/2018] [Indexed: 11/18/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is believed to be crucial for primary tumors to escape their original residence and invade and metastasize. To properly define EMT, there is a need for ligands that can identify this phenomenon in tumor tissue and invivo. A phage-display selection screening was performed to select novel binding phage peptides for identification of EMT in breast cancer. Epithelial breast cancer cell line, MCF-7 was transformed to mesenchymal phenotype by TGF-β treatment and was used for selection. Breast fibroblasts were used for subtractive depletion and breast cancer metastatic cell lines MDA-MB-231, T47D-shNMI were used for specificity assay. The binding peptides were identified, and their binding capacities were confirmed by phage capture assay, phage-based ELISA, immunofluorescence microscopy. The phage peptide bearing the 7-amino acid sequence, LGLRGSL, demonstrated selective binding to EMT phenotypic cells (MCF-7/TGF-β and MDA-MB-231) as compared to epithelial subtype, MCF-7, T47D and breast fibroblasts (Hs578T). The selected phage was also able to identify metastatic breast cancer tumor in breast cancer tissue microarray (TMA). These studies suggest that the selected phage peptide LGLRGSL identified by phage-display library, showed significant ability to bind to mesenchymal-like breast cancer cells/ tissues and can serve as a novel probe/ligand for metastatic breast cancer diagnostic and imaging.
Collapse
Affiliation(s)
- Kelvin M Jones
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States
| | - Balasubramanyam Karanam
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, United States
| | | | - Maninder Sandey
- Department of Pathobiology, Auburn University, Auburn, AL, United States
| | - Henry J Henderson
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States
| | - Rajeev S Samant
- Department of Pathobiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Samuel Temesgen
- Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States
| | - Clayton Yates
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, United States
| | - Deepa Bedi
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States
| |
Collapse
|
10
|
Li J, Li SH, Wu J, Weisel RD, Yao A, Stanford WL, Liu SM, Li RK. Young Bone Marrow Sca-1 Cells Rejuvenate the Aged Heart by Promoting Epithelial-to-Mesenchymal Transition. Am J Cancer Res 2018; 8:1766-1781. [PMID: 29556355 PMCID: PMC5858499 DOI: 10.7150/thno.22788] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/13/2018] [Indexed: 01/13/2023] Open
Abstract
Background: To improve the regenerative capacity of aged individuals, we reconstituted bone marrow (BM) of aged mice with young Sca-1 cells, which repopulated cardiac progenitors and prevented cardiac dysfunction after a myocardial infarction (MI). However, the mechanisms involved were incompletely elucidated. This study aimed to investigate whether young, highly regenerative BM Sca-1 cells exert their cardio-protective effects on the aged heart through reactivation of the epithelial-to-mesenchymal transition (EMT) process. Methods:In vitro, BM Sca-1 cells were co-cultured with epicardial-derived cells (EPDCs) under hypoxia condition; mRNA and protein levels of EMT genes were measured along with cellular proliferation and migration. In vivo, BM Sca-1+ or Sca-1- cells from young mice (2-3 months) were transplanted into lethally-irradiated old mice (20-22 months) to generate chimeras. In addition, Sca-1 knockout (KO) mice were reconstituted with wild type (WT) BM Sca-1+ cells. The effects of BM Sca-1 cell on EMT reactivation and improvement of cardiac function after MI were evaluated. Results:In vitro, BM Sca-1+ cells increased EPDC proliferation, migration, and EMT relative to Sca-1- cells and these effects were inhibited by a TGF-β blocker. In vivo, more young BM Sca-1+ than Sca-1- cells homed to the epicardium and induced greater host EPDC proliferation, migration, and EMT after MI. Furthermore, reconstitution of Sca-1 KO mice with WT Sca-1+ cells was associated with the reactivation of EMT and improved cardiac function after MI. Conclusions: Young BM Sca-1+ cells improved cardiac regeneration through promoting EPDC proliferation, migration and reactivation of EMT via the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Jiao Li
- Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada,Division of Cardiac Surgery, Department of Surgery, University of Toronto; Toronto, Canada
| | - Shu-Hong Li
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada
| | - Jun Wu
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada
| | - Richard D. Weisel
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada,Division of Cardiac Surgery, Department of Surgery, University of Toronto; Toronto, Canada
| | - Alina Yao
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada
| | - William L. Stanford
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa
| | - Shi-Ming Liu
- Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,✉ Corresponding author: Shi-Ming Liu, MD, Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China. Tel: 86-020-34153522; Fax: 86-20-3415-3709; and Ren-Ke Li, MD, PhD, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, Canada M5G 1L7. Tel: 1-416-581-7492; Fax: 1-416-581-7493;
| | - Ren-Ke Li
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada,Division of Cardiac Surgery, Department of Surgery, University of Toronto; Toronto, Canada,✉ Corresponding author: Shi-Ming Liu, MD, Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China. Tel: 86-020-34153522; Fax: 86-20-3415-3709; and Ren-Ke Li, MD, PhD, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, Canada M5G 1L7. Tel: 1-416-581-7492; Fax: 1-416-581-7493;
| |
Collapse
|
11
|
An androgen response element driven reporter assay for the detection of androgen receptor activity in prostate cells. PLoS One 2017; 12:e0177861. [PMID: 28570625 PMCID: PMC5453475 DOI: 10.1371/journal.pone.0177861] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/20/2017] [Indexed: 01/20/2023] Open
Abstract
The androgen receptor (AR) transcription factor plays a key role in the development and progression of prostate cancer, as is evident from the efficacy of androgen-deprivation therapy, AR is also the most frequently mutated gene, in castration resistant prostate cancer (CRPC). AR has therefore become an even more attractive therapeutic target in aggressive and disseminated prostate cancer. To investigate mechanisms of AR and AR target gene activation in different subpopulations of prostate cancer cells, a toolkit of AR expressor and androgen response element (ARE) reporter vectors were developed. Three ARE reporter vectors were constructed with different ARE consensus sequences in promoters linked to either fluorescence or luciferase reporter genes in lentiviral vector backbones. Cell lines transduced with the different vectors expressed the reporters in an androgen-dependent way according to fluorescence microscopy, flow cytometry and multi-well fluorescent and luminescence assays. Interestingly, the background reporter activity in androgen-depleted medium was significantly higher in LNCaP cells compared to the prostate transit amplifying epithelial cell lines, EP156T-AR and 957E/hTERT-AR with exogenous AR. The androgen-induced signal to background was much higher in the latter benign prostate cells than in LNCaP cells. Androgen-independent nuclear localization of AR was seen in LNCaP cells and reduced ARE-signaling was seen following treatment with abiraterone, an androgen synthesis inhibitor. The ARE reporter activity was significantly stronger when stimulated by androgens than by β-estradiol, progesterone and dexamethasone in all tested cell types. Finally, no androgen-induced ARE reporter activity was observed in tumorigenic mesenchymal progeny cells of EP156T cells following epithelial to mesenchymal transition. This underscores the observation that expression of the classical luminal differentiation transcriptome is restricted in mesenchymal type cells with or without AR expression, and presence of androgen.
Collapse
|
12
|
Axitinib blocks Wnt/β-catenin signaling and directs asymmetric cell division in cancer. Proc Natl Acad Sci U S A 2016; 113:9339-44. [PMID: 27482107 DOI: 10.1073/pnas.1604520113] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Oncogenic mutations of the Wnt (wingless)/β-catenin pathway are frequently observed in major cancer types. Thus far, however, no therapeutic agent targeting Wnt/β-catenin signaling is available for clinical use. Here we demonstrate that axitinib, a clinically approved drug, strikingly blocks Wnt/β-catenin signaling in cancer cells, zebrafish, and Apc(min/+) mice. Notably, axitinib dramatically induces Wnt asymmetry and nonrandom DNA segregation in cancer cells by promoting nuclear β-catenin degradation independent of the GSK3β (glycogen synthase kinase3β)/APC (adenomatous polyposis coli) complex. Using a DARTS (drug affinity-responsive target stability) assay coupled to 2D-DIGE (2D difference in gel electrophoresis) and mass spectrometry, we have identified the E3 ubiquitin ligase SHPRH (SNF2, histone-linker, PHD and RING finger domain-containing helicase) as the direct target of axitinib in blocking Wnt/β-catenin signaling. Treatment with axitinib stabilizes SHPRH and thereby increases the ubiquitination and degradation of β-catenin. Our findings suggest a previously unreported mechanism of nuclear β-catenin regulation and indicate that axitinib, a clinically approved drug, would provide therapeutic benefits for cancer patients with aberrant nuclear β-catenin activation.
Collapse
|
13
|
Olsen JR, Azeem W, Hellem MR, Marvyin K, Hua Y, Qu Y, Li L, Lin B, Ke XS, Øyan AM, Kalland KH. Context dependent regulatory patterns of the androgen receptor and androgen receptor target genes. BMC Cancer 2016; 16:377. [PMID: 27378372 PMCID: PMC4932678 DOI: 10.1186/s12885-016-2453-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 06/23/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Expression of the androgen receptor (AR) is associated with androgen-dependent proliferation arrest and terminal differentiation of normal prostate epithelial cells. Additionally, activation of the AR is required for survival of benign luminal epithelial cells and primary cancer cells, thus androgen deprivation therapy (ADT) leads to apoptosis in both benign and cancerous tissue. Escape from ADT is known as castration-resistant prostate cancer (CRPC). In the course of CRPC development the AR typically switches from being a cell-intrinsic inhibitor of normal prostate epithelial cell proliferation to becoming an oncogene that is critical for prostate cancer cell proliferation. A clearer understanding of the context dependent activation of the AR and its target genes is therefore desirable. METHODS Immortalized human prostate basal epithelial EP156T cells and progeny cells that underwent epithelial to mesenchymal transition (EMT), primary prostate epithelial cells (PrECs) and prostate cancer cell lines LNCaP, VCaP and 22Rv1 were used to examine context dependent restriction and activation of the AR and classical target genes, such as KLK3. Genome-wide gene expression analyses and single cell protein analyses were applied to study the effect of different contexts. RESULTS A variety of growth conditions were tested and found unable to activate AR expression and transcription of classical androgen-dependent AR target genes, such as KLK3, in prostate epithelial cells with basal cell features or in mesenchymal type prostate cells. The restriction of androgen- and AR-dependent transcription of classical target genes in prostate basal epithelial cells was at the level of AR expression. Exogenous AR expression was sufficient for androgen-dependent transcription of AR target genes in prostate basal epithelial cells, but did not exert a positive feedback on endogenous AR expression. Treatment of basal prostate epithelial cells with inhibitors of epigenetic gene silencing was not efficient in inducing androgen-dependent transcription of AR target genes, suggesting the importance of missing cofactor(s). CONCLUSIONS Regulatory mechanisms of AR and androgen-dependent AR target gene transcription are insufficiently understood and may be critical for prostate cancer initiation, progression and escape from standard therapy. The present model is useful for the study of context dependent activation of the AR and its transcriptome.
Collapse
Affiliation(s)
- Jan Roger Olsen
- Department of Clinical Science, University of Bergen, Bergen, Norway. .,, Laboratory Bld. 5. etg, Bergen Health, Bergen, NO-5021, Norway.
| | - Waqas Azeem
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | | | - Kristo Marvyin
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Yaping Hua
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Yi Qu
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Lisha Li
- Cancer Institute, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Biaoyang Lin
- Cancer Institute, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Department of Urology, University of Washington, Seattle, WA, USA
| | - Xi- Song Ke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Karl- Henning Kalland
- Department of Clinical Science, University of Bergen, Bergen, Norway. .,Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway. .,Department of Microbiology, Haukeland University Hospital, Bergen, Norway. .,, Laboratory Bld. 5. etg, Bergen Health, Bergen, NO-5021, Norway.
| |
Collapse
|
14
|
Reigstad I, Smeland HYH, Skogstrand T, Sortland K, Schmid MC, Reed RK, Stuhr L. Stromal Integrin α11β1 Affects RM11 Prostate and 4T1 Breast Xenograft Tumors Differently. PLoS One 2016; 11:e0151663. [PMID: 26990302 PMCID: PMC4798484 DOI: 10.1371/journal.pone.0151663] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/02/2016] [Indexed: 11/18/2022] Open
Abstract
PURPOSE It has been implied that the collagen binding integrin α11β1 plays a role in carcinogenesis. As still relatively little is known about how the stromal integrin α11β1 affects different aspects of tumor development, we wanted to examine the direct effects on primary tumor growth, fibrosis, tumor interstitial fluid pressure (PIF) and metastasis in murine 4T1 mammary and RM11 prostate tumors, using an in vivo SCID integrin α11-deficient mouse model. METHODS Tumor growth was measured using a caliper, PIF by the wick-in-needle technique, activated fibroblasts by α-SMA immunofluorescence staining and fibrosis by transmission electron microscopy and picrosirius-red staining. Metastases were evaluated using hematoxylin and eosin stained sections. RESULTS RM11 tumor growth was significantly reduced in the SCID integrin α11-deficient (α11-KO) compared to in SCID integrin α11 wild type (WT) mice, whereas there was no similar effect in the 4T1 tumor model. The 4T1 model demonstrated an alteration in collagen fibril diameter in the integrin α11-KO mice compared to WT, which was not found in the RM11 model. There were no significant differences in the amount of activated fibroblasts, total collagen content, collagen organization or PIF in the tumors in integrin α11-deficient mice compared to WT mice. There was also no difference in lung metastases between the two groups. CONCLUSION Deficiency of stromal integrin α11β1 showed different effects on tumor growth and collagen fibril diameter depending on tumor type, but no effect on tumor PIF or development of lung metastasis.
Collapse
Affiliation(s)
- Inga Reigstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Matrix biology group, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| | - Hilde Y. H. Smeland
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Center of Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Trude Skogstrand
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Matrix biology group, Haukeland University Hospital, Bergen, Norway
| | - Kristina Sortland
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Matrix biology group, Haukeland University Hospital, Bergen, Norway
| | - Marei Caroline Schmid
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Center of Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Rolf K. Reed
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Center of Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Linda Stuhr
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
15
|
Zeltz C, Gullberg D. The integrin-collagen connection--a glue for tissue repair? J Cell Sci 2016; 129:653-64. [PMID: 26857815 DOI: 10.1242/jcs.180992] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The α1β1, α2β1, α10β1 and α11β1 integrins constitute a subset of the integrin family with affinity for GFOGER-like sequences in collagens. Integrins α1β1 and α2β1 were originally identified on a subset of activated T-cells, and have since been found to be expressed on a number of cell types including platelets (α2β1), vascular cells (α1β1, α2β1), epithelial cells (α1β1, α2β1) and fibroblasts (α1β1, α2β1). Integrin α10β1 shows a distribution that is restricted to mesenchymal stem cells and chondrocytes, whereas integrin α11β1 appears restricted to mesenchymal stem cells and subsets of fibroblasts. The bulk of the current literature suggests that collagen-binding integrins only have a limited role in adult connective tissue homeostasis, partly due to a limited availability of cell-binding sites in the mature fibrillar collagen matrices. However, some recent data suggest that, instead, they are more crucial for dynamic connective tissue remodeling events--such as wound healing--where they might act specifically to remodel and restore the tissue architecture. This Commentary discusses the recent development in the field of collagen-binding integrins, their roles in physiological and pathological settings with special emphasis on wound healing, fibrosis and tumor-stroma interactions, and include a discussion of the most recently identified newcomers to this subfamily--integrins α10β1 and α11β1.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| |
Collapse
|
16
|
Yang K, Wu WM, Chen YC, Lo SH, Liao YC. ΔNp63α Transcriptionally Regulates the Expression of CTEN That Is Associated with Prostate Cell Adhesion. PLoS One 2016; 11:e0147542. [PMID: 26784942 PMCID: PMC4718700 DOI: 10.1371/journal.pone.0147542] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 01/04/2016] [Indexed: 01/02/2023] Open
Abstract
p63 is a member of the p53 transcription factor family and a linchpin of epithelial development and homeostasis. p63 drives the expression of many target genes involved in cell survival, adhesion, migration and cancer. In this study, we identify C-terminal tensin-like (CTEN) molecule as a downstream target of ΔNp63α, the predominant p63 isoform expressed in epithelium. CTEN belongs to the tensin family and is mainly localized to focal adhesions, which mediate many biological events such as cell adhesion, migration, proliferation and gene expression. Our study demonstrate that ΔNp63 and CTEN are both highly expressed in normal prostate epithelial cells and are down-regulated in prostate cancer. In addition, reduced expression of CTEN and ΔNp63 is correlated with prostate cancer progression from primary tumors to metastatic lesions. Silencing of ΔNp63 leads to decreased mRNA and protein levels of CTEN. ΔNp63α induces transcriptional activity of the CTEN promoter and a 140-bp fragment upstream of the transcription initiation site is the minimal promoter region required for activation. A putative binding site for p63 is located between -61 and -36 within the CTEN promoter and mutations of the critical nucleotides in this region abolish ΔNp63α-induced promoter activity. The direct interaction of ΔNp63α with the CTEN promoter was demonstrated using a chromatin immunoprecipitation (ChIP) assay. Moreover, impaired cell adhesion caused by ΔNp63α depletion is rescued by over-expression of CTEN, suggesting that CTEN is a downstream effector of ΔNp63α-mediated cell adhesion. In summary, our findings demonstrate that ΔNp63α functions as a trans-activation factor of CTEN promoter and regulates cell adhesion through modulating CTEN. Our study further contributes to the potential regulatory mechanisms of CTEN in prostate cancer progression.
Collapse
Affiliation(s)
- Kuan Yang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Wei-Ming Wu
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Ya-Chi Chen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Su Hao Lo
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, California, United States of America
| | - Yi-Chun Liao
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
17
|
Tu H, Huhtala P, Lee HM, Adams JC, Pihlajaniemi T. Membrane-associated collagens with interrupted triple-helices (MACITs): evolution from a bilaterian common ancestor and functional conservation in C. elegans. BMC Evol Biol 2015; 15:281. [PMID: 26667623 PMCID: PMC4678570 DOI: 10.1186/s12862-015-0554-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 12/02/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Collagens provide structural support and guidance cues within the extracellular matrix of metazoans. Mammalian collagens XIII, XXIII and XXV form a unique subgroup of type II transmembrane proteins, each comprising a short N-terminal cytosolic domain, a transmembrane domain and a largely collagenous ectodomain. We name these collagens as MACITs (Membrane-Associated Collagens with Interrupted Triple-helices), and here investigate their evolution and conserved properties. To date, these collagens have been studied only in mammals. Knowledge of the representation of MACITs in other extant metazoans is lacking. This question is of interest for understanding structural/functional relationships in the MACIT family and also for insight into the evolution of MACITs in relation to the secreted, fibrillar collagens that are present throughout the metazoa. RESULTS MACITs are restricted to bilaterians and are represented in the Ecdysozoa, Hemichordata, Urochordata and Vertebrata (Gnathostomata). They were not identified in available early-diverging metazoans, Lophotrochozoa, Echinodermata, Cephalochordata or Vertebrata (Cyclostomata). Whereas invertebrates encode a single MACIT, collagens XIII/XXIII/XXV of jawed vertebrates are paralogues that originated from the two rounds of en-bloc genome duplication occurring early in vertebrate evolution. MACITs have conserved domain architecture in which a juxta-membrane furin-cleavage site and the C-terminal 34 residues are especially highly conserved, whereas the cytoplasmic domains are weakly conserved. To study protein expression and function in a metazoan with a single MACIT gene, we focused on Caenorhabditis elegans and its col-99 gene. A col-99 cDNA was cloned and expressed as protein in mammalian CHO cells, two antibodies against COL-99 protein were generated, and a col-99-bearing fosmid gene construct col-99::egfp::flag was used to generate transgenic C. elegans lines. The encoded COL-99 polypeptide is 85 kDa in size and forms a trimeric protein. COL-99 is plasma membrane-associated and undergoes furin-dependent ectodomain cleavage and shedding. COL-99 is detected in mouth, pharynx, body wall and the tail, mostly in motor neurons and muscle systems and is enriched at neuromuscular junctions. CONCLUSIONS Through identification of MACITs in multiple metazoan phyla we developed a model for the evolution of MACITs. The experimental data demonstrate conservation of MACIT molecular and cellular properties and tissue localisations in the invertebrate, C. elegans.
Collapse
Affiliation(s)
- Hongmin Tu
- Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Aapistie 5, Oulu, FIN 90014, Finland.
| | - Pirkko Huhtala
- Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Aapistie 5, Oulu, FIN 90014, Finland.
| | - Hang-Mao Lee
- Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Aapistie 5, Oulu, FIN 90014, Finland.
| | - Josephine C Adams
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| | - Taina Pihlajaniemi
- Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Aapistie 5, Oulu, FIN 90014, Finland.
| |
Collapse
|
18
|
Drivalos A, Chrisofos M, Efstathiou E, Kapranou A, Kollaitis G, Koutlis G, Antoniou N, Karanastasis D, Dimopoulos MA, Bamias A. Expression of α5-integrin, α7-integrin, Ε-cadherin, and N-cadherin in localized prostate cancer. Urol Oncol 2015; 34:165.e11-8. [PMID: 26652134 DOI: 10.1016/j.urolonc.2015.10.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 07/14/2015] [Accepted: 10/27/2015] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To explore the correlation between the expression of α5-integrin, α7-integrin, Ε-cadherin, and N-cadherin in prostate cancer (PCa) and its clinicopathological data including tumor grade and clinical stage. METHODS The expression of α5-integrin, α7-integrin, Ε-cadherin, and N-cadherin was examined in 157 cases of PCa and adjacent normal prostatic tissue by immunohistochemical assay, and the correlation with clinicopathological features was analyzed. RESULTS Expressions of α5-integrin, α7-integrin, and Ε-cadherin in PCa were lower than those in normal prostatic tissues (P<0.05). N-cadherin expression was higher in cancer prostatic tissue than in normal prostatic tissues (P<0.05). The reduced expression of α5-integrin, α7-integrin, and Ε-cadherin was related to Gleason score, pathological stage, lymph node metastasis, and prostate-specific antigen level, but it was not associated with positive surgical margins and patient age. The increased expression of N-cadherin was related to Gleason score, pathological stage, lymph node metastasis, and prostate-specific antigen level, but not to age and positive surgical margins. The expression of E-cadherin was highly negatively correlated with that of N-cadherin and also positively correlated with that of α5-integrin and α7-integrin. CONCLUSION The reduced expression of α5-integrin, α7-integrin, and Ε-cadherin and abnormal expression of N-cadherin play an important role in the occurrence and development of PCa. The results indicate that these have potential values in the diagnosis and are predictable indices in the proliferation of PCa.
Collapse
Affiliation(s)
| | - Michael Chrisofos
- 2nd Department of Urology, School of Medicine, Sismanoglio Hospital, University of Athens, Athens, Greece
| | - Eleni Efstathiou
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, University of Athens, Athens, Greece
| | - Amalia Kapranou
- Department of Anatomopathology, Athens Navy Hospital, Athens, Greece
| | | | - Georgios Koutlis
- Department of Anatomopathology, Athens Navy Hospital, Athens, Greece
| | - Nick Antoniou
- Department of Urology, Athens General Hospital "Elpis," Athens, Greece
| | | | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, University of Athens, Athens, Greece
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, University of Athens, Athens, Greece
| |
Collapse
|
19
|
Abstract
The paper gives general information about the epithelial-mesenchymal transition (EMT), its morphological manifestations, altered expression of a number of proteins, types of EMT, and its role in embryogenesis and human diseases, including that about EMT as a mechanism by which the tumor cell acquires prometastatic potential. EMT is a process that is essential in health for gastrulation and the formation of neural crest cells; however, it is also important for the development of abnormalities, among other processes, organ fibrosis and tumor metastases. An understanding of the role of EMT in cancer spread has led to active studies of the process in the past decades. Despite the fact that there are sufficiently many publications on different aspects of EMT, the exact mechanisms regulating the process and the possibility for its therapeutic exposure remain unclear. There is also evidence on the possible association of EMT with the generation of cancer stem cells in tumors.
Collapse
Affiliation(s)
- M V Puchinskaya
- Belarusian State Medical University, Minsk, Republic of Belarus
| |
Collapse
|
20
|
Bitting RL, Schaeffer D, Somarelli JA, Garcia-Blanco MA, Armstrong AJ. The role of epithelial plasticity in prostate cancer dissemination and treatment resistance. Cancer Metastasis Rev 2014; 33:441-68. [PMID: 24414193 PMCID: PMC4230790 DOI: 10.1007/s10555-013-9483-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nearly 30,000 men die annually in the USA of prostate cancer, nearly uniformly from metastatic dissemination. Despite recent advances in hormonal, immunologic, bone-targeted, and cytotoxic chemotherapies, treatment resistance and further dissemination are inevitable in men with metastatic disease. Emerging data suggests that the phenomenon of epithelial plasticity, encompassing both reversible mesenchymal transitions and acquisition of stemness traits, may underlie this lethal biology of dissemination and treatment resistance. Understanding the molecular underpinnings of this cellular plasticity from preclinical models of prostate cancer and from biomarker studies of human metastatic prostate cancer has provided clues to novel therapeutic approaches that may delay or prevent metastatic disease and lethality over time. This review will discuss the preclinical and clinical evidence for epithelial plasticity in this rapidly changing field and relate this to clinical phenotype and resistance in prostate cancer while suggesting novel therapeutic approaches.
Collapse
Affiliation(s)
- Rhonda L. Bitting
- Division of Medical Oncology, Duke Cancer Institute, Duke University, DUMC Box 102002, Durham, NC 27710, USA. Department of Medicine, Duke University, Durham, NC, USA. Center for RNA Biology, Duke University, Durham, NC, USA
| | - Daneen Schaeffer
- Center for RNA Biology, Duke University, Durham, NC, USA. Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Jason A. Somarelli
- Center for RNA Biology, Duke University, Durham, NC, USA. Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Mariano A. Garcia-Blanco
- Department of Medicine, Duke University, Durham, NC, USA. Center for RNA Biology, Duke University, Durham, NC, USA. Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Andrew J. Armstrong
- Division of Medical Oncology, Duke Cancer Institute, Duke University, DUMC Box 102002, Durham, NC 27710, USA. Department of Medicine, Duke University, Durham, NC, USA. Center for RNA Biology, Duke University, Durham, NC, USA. Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| |
Collapse
|
21
|
Gaitas A, Malhotra R, Pienta K, Kim G. Response to "Comment on 'A method to measure cellular adhesion utilizing a polymer micro-cantilever'" [Appl. Phys. Lett. 104, 236103 (2014)]. APPLIED PHYSICS LETTERS 2014; 104:236104. [PMID: 25315106 PMCID: PMC4187251 DOI: 10.1063/1.4882185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/28/2014] [Indexed: 06/04/2023]
Affiliation(s)
- Angelo Gaitas
- PicoCal, Inc., 333 Parkland Plaza, Ann Arbor, Michigan 48103, USA
| | - Ricky Malhotra
- PicoCal, Inc., 333 Parkland Plaza, Ann Arbor, Michigan 48103, USA
| | - Kenneth Pienta
- Department of Urology, Johns Hopkins University School of Medicine , Marburg 121, 600 N. Wolfe Street, Baltimore, Maryland 21287-2101, USA
| | - Gwangseong Kim
- PicoCal, Inc., 333 Parkland Plaza, Ann Arbor, Michigan 48103, USA
| |
Collapse
|
22
|
Zeltz C, Lu N, Gullberg D. Integrin α11β1: A Major Collagen Receptor on Fibroblastic Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 819:73-83. [DOI: 10.1007/978-94-017-9153-3_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
23
|
Klein K, Rommel CE, Hirschfeld-Warneken VC, Spatz JP. Cell membrane topology analysis by RICM enables marker-free adhesion strength quantification. Biointerphases 2013; 8:28. [DOI: 10.1186/1559-4106-8-28] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/11/2013] [Indexed: 11/10/2022] Open
|
24
|
Qu Y, Oyan AM, Liu R, Hua Y, Zhang J, Hovland R, Popa M, Liu X, Brokstad KA, Simon R, Molven A, Lin B, Zhang WD, McCormack E, Kalland KH, Ke XS. Generation of prostate tumor-initiating cells is associated with elevation of reactive oxygen species and IL-6/STAT3 signaling. Cancer Res 2013; 73:7090-100. [PMID: 24101153 DOI: 10.1158/0008-5472.can-13-1560] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
How prostate cancer is initiated remains a topic of debate. In an effort to establish a human model of prostate carcinogenesis, we adapted premalignant human prostate EPT2-D5 cells to protein-free medium to generate numerous tight prostate spheres (D5HS) in monolayer culture. In contrast to EPT2-D5 cells, the newly generated D5HS efficiently formed large subcutaneous tumors and subsequent metastases in vivo, showing the tumorigenicity of D5HS spheres. A striking production of interleukin (IL)-6 mRNA and protein was found in D5HS cells. The essential roles of IL-6 and the downstream STAT3 signaling in D5HS tumor sphere formation were confirmed by neutralizing antibody, chemical inhibitors, and fluorescent pathway reporter. In addition, elevated reactive oxygen species (ROS) produced upon protein depletion was required for the activation of IL-6/STAT3 in D5HS. Importantly, a positive feedback loop was found between ROS and IL-6 during tumor sphere formation. The association of ROS/IL-6/STAT3 to the carcinogenesis of human prostate cells was further examined in xenograft tumors and verified by limiting dilution implantations. Collectively, we have for the first time established human prostate tumor-initiating cells based on physiologic adaption. The intrinsic association of ROS and IL-6/STAT3 signaling in human prostate carcinogenesis shed new light on this relationship and define therapeutic targets in this setting.
Collapse
Affiliation(s)
- Yi Qu
- Authors' Affiliations: The Gade Institute, Broegelmann Research Laboratory, Department of Clinical Science, Department of Medicine, University of Bergen; Departments of Microbiology and Pathology, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital; KinN Therapeutics AS, Bergen, Norway; College of Pharmacy, Second Military Medical University, Shanghai, PR China; Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Zhejiang-California International NanoSystems Institute, Zhejiang University, Hangzhou, PR China; Swedish Medical Center; and Department of Urology, University of Washington, Seattle, Washington
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gaitas A, Malhotra R, Pienta K. A method to measure cellular adhesion utilizing a polymer micro-cantilever. APPLIED PHYSICS LETTERS 2013; 103:123702. [PMID: 24170959 PMCID: PMC3790771 DOI: 10.1063/1.4821946] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/06/2013] [Indexed: 06/01/2023]
Abstract
In the present study we engineered a micro-machined polyimide cantilever with an embedded sensing element to investigate cellular adhesion, in terms of its relative ability to stick to a cross-linker, 3,3'-dithiobis[sulfosuccinimidylpropionate], coated on the cantilever surface. To achieve this objective, we investigated adhesive properties of three human prostate cancer cell lines, namely, a bone metastasis derived human prostate cancer cell line (PC3), a brain metastasis derived human prostate cancer cell line (DU145), and a subclone of PC3 (PC3-EMT14). We found that PC3-EMT14, which displays a mesenchymal phenotype, has the least adhesion compared to PC3 and DU145, which exhibit an epithelial phenotype.
Collapse
Affiliation(s)
- Angelo Gaitas
- PicoCal, Inc., 333 Parkland Plaza, Ann Arbor, Michigan 48103, USA ; Electronic Instrumentation Laboratory, TU Delft, Mekelweg 4, 2628CD Delft, The Netherlands
| | | | | |
Collapse
|
26
|
Reis STD, Leite KRM, Mosconi Neto A, Pontes Júnior J, Viana NI, Antunes AA, Dall'Oglio MF, Srougi M. Immune expression of E-cadherin and α, β and γ-Catenin adhesion molecules and prognosis for upper urinary tract urothelial carcinomas. Int Braz J Urol 2013; 38:466-73. [PMID: 22951176 DOI: 10.1590/s1677-55382012000400005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2012] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Cell adhesion molecules (CAM) are required for maintaining a normal epithelial phenotype, and abnormalities in CAM expression have been related to cancer progression, including bladder urothelial carcinomas. There is only one study that correlates E-cadherin and Α-, Β- and y-catenin expression with prognosis of upper tract urothelial carcinomas. Our aim is to study the pattern of immune expression of these CAMs in urothelial carcinomas from the renal pelvis and ureter in patients who have been treated surgically. Our goal is to correlate these expression levels and characteristics with well-known prognostic parameters for disease-free survival. MATERIALS AND METHODS We evaluated specimens from 20 patients with urothelial carcinomas of the renal pelvis and ureter who were treated with nephroureterectomy or ureterectomy between June 1997 and January 2007. CAM expression was evaluated by immunohistochemistry in a tissue microarray and correlated with histopathological characteristics and patient outcomes after a mean follow-up of 55 months. RESULTS We observed a relationship between E-cadherin expression and disease recurrence. Disease recurrence occurred in 87.5% of patients with strong E-cadherin expression. Only 50.0% of patients with moderate expression and 0% of patients with weak or no expression of E-cadherin had disease recurrence (p = 0.014). There was also a difference in disease-free survival. Patients with strong E-cadherin expression had a mean disease-free survival rate of 49.1 months, compared to 83.9 months for patients with moderate expression (p = 0.011). Additionally, an absence of Α-catenin expression was associated with tumors that were larger than 3 cm (p = 0.003). CONCLUSIONS We demonstrated for the first time that immune expression of E-cadherin is related to tumor recurrence and disease-free survival rates, and the absence of Α-catenin expression is related to tumor size in upper tract urothelial carcinomas.
Collapse
Affiliation(s)
- Sabrina Thalita dos Reis
- Laboratory of Medical Investigation (LIM55), Urology Department, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
27
|
p63 attenuates epithelial to mesenchymal potential in an experimental prostate cell model. PLoS One 2013; 8:e62547. [PMID: 23658742 PMCID: PMC3641034 DOI: 10.1371/journal.pone.0062547] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 03/22/2013] [Indexed: 02/06/2023] Open
Abstract
The transcription factor p63 is central for epithelial homeostasis and development. In our model of epithelial to mesenchymal transition (EMT) in human prostate cells, p63 was one of the most down-regulated transcription factors during EMT. We therefore investigated the role of p63 in EMT. Over-expression of the predominant epithelial isoform ΔNp63α in mesenchymal type cells of the model led to gain of several epithelial characteristics without resulting in a complete mesenchymal to epithelial transition (MET). This was corroborated by a reciprocal effect when p63 was knocked down in epithelial EP156T cells. Global gene expression analyses showed that ΔNp63α induced gene modules involved in both cell-to-cell and cell-to-extracellular-matrix junctions in mesenchymal type cells. Genome-wide analysis of p63 binding sites using ChIP-seq analyses confirmed binding of p63 to regulatory areas of genes associated with cell adhesion in prostate epithelial cells. DH1 and ZEB1 are two elemental factors in the control of EMT. Over-expression and knock-down of these factors, respectively, were not sufficient alone or in combination with ΔNp63α to reverse completely the mesenchymal phenotype. The partial reversion of epithelial to mesenchymal transition might reflect the ability of ΔNp63α, as a key co-ordinator of several epithelial gene expression modules, to reduce epithelial to mesenchymal plasticity (EMP). The utility of ΔNp63α expression and the potential of reduced EMP in order to counteract metastasis warrant further investigation.
Collapse
|
28
|
Qu Y, Li WC, Hellem MR, Rostad K, Popa M, McCormack E, Oyan AM, Kalland KH, Ke XS. MiR-182 and miR-203 induce mesenchymal to epithelial transition and self-sufficiency of growth signals via repressing SNAI2 in prostate cells. Int J Cancer 2013; 133:544-55. [PMID: 23354685 DOI: 10.1002/ijc.28056] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 01/10/2013] [Indexed: 01/10/2023]
Abstract
MicroRNAs play critical roles in tumorigenesis and metastasis. Here, we report the dual functions of miR-182 and miR-203 in our previously described prostate cell model. MiR-182 and miR-203 were completely repressed during epithelial to mesenchymal transition (EMT) from prostate epithelial EP156T cells to the progeny mesenchymal nontransformed EPT1 cells. Re-expression of miR-182 or miR-203 in EPT1 cells and prostate cancer PC3 cells induced mesenchymal to epithelial transition (MET) features. Simultaneously, miR-182 and miR-203 provided EPT1 cells with the ability to self-sufficiency of growth signals, a well-recognized oncogenic feature. Gene expression profiling showed high overlap of the genes affected by miR-182 and miR-203. SNAI2 was identified as a common target of miR-182 and miR-203. Knock-down of SNAI2 in EPT1 cells phenocopied re-expression of either miR-182 or miR-203 regarding both MET and self-sufficiency of growth signals. Strikingly, considerable overlaps of changed genes were found between the re-expression of miR-182/203 and knock-down of SNAI2. Finally, P-cadherin was identified as a direct target of SNAI2. We conclude that miR-182 and miR-203 induce MET features and growth factor independent growth via repressing SNAI2 in prostate cells. Our findings shed new light on the roles of miR-182/203 in cancer related processes.
Collapse
Affiliation(s)
- Yi Qu
- The Gade Institute, University of Bergen, Bergen, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Klein K, Maier T, Hirschfeld-Warneken VC, Spatz JP. Marker-free phenotyping of tumor cells by fractal analysis of reflection interference contrast microscopy images. NANO LETTERS 2013; 13:5474-9. [PMID: 24079895 PMCID: PMC3831548 DOI: 10.1021/nl4030402] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 09/26/2013] [Indexed: 05/21/2023]
Abstract
Phenotyping of tumor cells by marker-free quantification is important for cancer diagnostics. For the first time, fractal analysis of reflection interference contrast microscopy images of single living cells was employed as a new method to distinguish between different nanoscopic membrane features of tumor cells. Since tumor progression correlates with a higher degree of chaos within the cell, it can be quantified mathematically by fractality. Our results show a high accuracy in identifying malignant cells with a failure chance of 3%, which is far better than today's applied methods.
Collapse
|
30
|
Armstrong SR, Narendrula R, Guo B, Parissenti AM, McCallum KL, Cull S, Lannér C. Distinct genetic alterations occur in ovarian tumor cells selected for combined resistance to carboplatin and docetaxel. J Ovarian Res 2012. [PMID: 23194409 PMCID: PMC3541348 DOI: 10.1186/1757-2215-5-40] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
UNLABELLED BACKGROUND Current protocols for the treatment of ovarian cancer include combination chemotherapy with a platinating agent and a taxane. However, many patients experience relapse of their cancer and the development of drug resistance is not uncommon, making successful second line therapy difficult to achieve. The objective of this study was to develop and characterize a cell line resistant to both carboplatin and docetaxel (dual drug resistant ovarian cell line) and to compare this cell line to cells resistant to either carboplatin or docetaxel. METHODS The A2780 epithelial endometrioid ovarian cancer cell line was used to select for isogenic carboplatin, docetaxel and dual drug resistant cell lines. A selection method of gradually increasing drug doses was implemented to avoid clonal selection. Resistance was confirmed using a clonogenic assay. Changes in gene expression associated with the development of drug resistance were determined by microarray analysis. Changes in the expression of selected genes were validated by Quantitative Real-Time Polymerase Chain Reaction (QPCR) and immunoblotting. RESULTS Three isogenic cell lines were developed and resistance to each drug or the combination of drugs was confirmed. Development of resistance was accompanied by a reduced growth rate. The microarray and QPCR analyses showed that unique changes in gene expression occurred in the dual drug resistant cell line and that genes known to be involved in resistance could be identified in all cell lines. CONCLUSIONS Ovarian tumor cells can acquire resistance to both carboplatin and docetaxel when selected in the presence of both agents. Distinct changes in gene expression occur in the dual resistant cell line indicating that dual resistance is not a simple combination of the changes observed in cell lines exhibiting single agent resistance.
Collapse
|
31
|
Bünger S, Barow M, Thorns C, Freitag-Wolf S, Danner S, Tiede S, Pries R, Görg S, Bruch HP, Roblick U, Kruse C, Habermann J. Pancreatic Carcinoma Cell Lines Reflect Frequency and Variability of Cancer Stem Cell Markers in Clinical Tissue. Eur Surg Res 2012; 49:88-98. [DOI: 10.1159/000341669] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 07/05/2012] [Indexed: 12/20/2022]
|
32
|
Baek ST, Tallquist MD. Nf1 limits epicardial derivative expansion by regulating epithelial to mesenchymal transition and proliferation. Development 2012; 139:2040-9. [PMID: 22535408 DOI: 10.1242/dev.074054] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The epicardium is the primary source of coronary vascular smooth muscle cells (cVSMCs) and fibroblasts that reside in the compact myocardium. To form these epicardial-derived cells (EPDCs), the epicardium undergoes the process of epithelial to mesenchymal transition (EMT). Although several signaling pathways have been identified that disrupt EMT, no pathway has been reported that restricts this developmental process. Here, we identify neurofibromin 1 (Nf1) as a key mediator of epicardial EMT. To determine the function of Nf1 during epicardial EMT and the formation of epicardial derivatives, cardiac fibroblasts and cVSMCs, we generated mice with a tissue-specific deletion of Nf1 in the epicardium. We found that mutant epicardial cells transitioned more readily to mesenchymal cells in vitro and in vivo. The mesothelial epicardium lost epithelial gene expression and became more invasive. Using lineage tracing of EPDCs, we found that the process of EMT occurred earlier in Nf1 mutant hearts, with an increase in epicardial cells entering the compact myocardium. Moreover, loss of Nf1 caused increased EPDC proliferation and resulted in more cardiac fibroblasts and cVSMCs. Finally, we were able to partially reverse the excessive EMT caused by loss of Nf1 by disrupting Pdgfrα expression in the epicardium. Conversely, Nf1 activation was able to inhibit PDGF-induced epicardial EMT. Our results demonstrate a regulatory role for Nf1 during epicardial EMT and provide insights into the susceptibility of patients with disrupted NF1 signaling to cardiovascular disease.
Collapse
Affiliation(s)
- Seung Tae Baek
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | | |
Collapse
|
33
|
Celià-Terrassa T, Meca-Cortés O, Mateo F, Martínez de Paz A, Rubio N, Arnal-Estapé A, Ell BJ, Bermudo R, Díaz A, Guerra-Rebollo M, Lozano JJ, Estarás C, Ulloa C, Álvarez-Simón D, Milà J, Vilella R, Paciucci R, Martínez-Balbás M, de Herreros AG, Gomis RR, Kang Y, Blanco J, Fernández PL, Thomson TM. Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells. J Clin Invest 2012; 122:1849-68. [PMID: 22505459 DOI: 10.1172/jci59218] [Citation(s) in RCA: 362] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 02/29/2012] [Indexed: 12/15/2022] Open
Abstract
Malignant progression in cancer requires populations of tumor-initiating cells (TICs) endowed with unlimited self renewal, survival under stress, and establishment of distant metastases. Additionally, the acquisition of invasive properties driven by epithelial-mesenchymal transition (EMT) is critical for the evolution of neoplastic cells into fully metastatic populations. Here, we characterize 2 human cellular models derived from prostate and bladder cancer cell lines to better understand the relationship between TIC and EMT programs in local invasiveness and distant metastasis. The model tumor subpopulations that expressed a strong epithelial gene program were enriched in highly metastatic TICs, while a second subpopulation with stable mesenchymal traits was impoverished in TICs. Constitutive overexpression of the transcription factor Snai1 in the epithelial/TIC-enriched populations engaged a mesenchymal gene program and suppressed their self renewal and metastatic phenotypes. Conversely, knockdown of EMT factors in the mesenchymal-like prostate cancer cell subpopulation caused a gain in epithelial features and properties of TICs. Both tumor cell subpopulations cooperated so that the nonmetastatic mesenchymal-like prostate cancer subpopulation enhanced the in vitro invasiveness of the metastatic epithelial subpopulation and, in vivo, promoted the escape of the latter from primary implantation sites and accelerated their metastatic colonization. Our models provide new insights into how dynamic interactions among epithelial, self-renewal, and mesenchymal gene programs determine the plasticity of epithelial TICs.
Collapse
Affiliation(s)
- Toni Celià-Terrassa
- Department of Cell Biology, Barcelona Institute of Molecular Biology, Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Pontes-Junior J, Reis ST, Dall'Oglio M, Neves de Oliveira LC, Cury J, Carvalho PA, Ribeiro-Filho LA, Moreira Leite KR, Srougi M. Evaluation of the expression of integrins and cell adhesion molecules through tissue microarray in lymph node metastases of prostate cancer. J Carcinog 2011; 8:3. [PMID: 19240373 PMCID: PMC2678866 DOI: 10.4103/1477-3163.48453] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Integrins and adhesion molecules are responsible for the maintenance of the epithelial phenotype. Cell culture studies have reported the correlation between adhesion molecule expression and prostate carcinoma, but their role in the metastatic process is not yet known. Our aim is to study the expression profiles of these molecules and evaluate their association with the metastatic behavior of prostate adenocarcinoma. Materials and Methods: A Tissue Microarray containing two samples from 19 primary tumors and one from their corresponding lymph node metastases was constructed and subjected to immunohistochemical analysis of the expression of integrins, E-cadherin and β and γ-catenins. Within each case, paired analyses were also performed to evaluate gains or losses in metastasis compared to its primary tumor. Results: The expression of αv, αvβ3, α2β1 and γ-catenin were abnormal in almost every case. Marked loss of E-cadherin and β4 integrin was found in primary and metastatic lesions. β-catenin was normal in all primary cases and in 94% of metastases. α6 was normal in all primary tumors and metastases. α3 and α3β1 were normal in 32% of primary cases and in 53% and 6% of metastases, respectively. In paired analyses, loss of E-cadherin, β4, αv, α3β1 and αvβ3 was found in 65%, 71%, 59%, 53% and 47% of patients, respectively. Catenins and α2β1 showed maintenance of expression in most of the cases. Conclusions: In this preliminary study we have shown that the loss of cell adhesion molecules can be considered a characteristic of the metastatic phenotype in prostate cancer. Larger series should be evaluated in order to confirm our findings.
Collapse
Affiliation(s)
- Jose Pontes-Junior
- Laboratory of Medical Investigation - LIM 55, Urology Department, Medical School University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
TMPRSS2/ERG promotes epithelial to mesenchymal transition through the ZEB1/ZEB2 axis in a prostate cancer model. PLoS One 2011; 6:e21650. [PMID: 21747944 PMCID: PMC3128608 DOI: 10.1371/journal.pone.0021650] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 06/04/2011] [Indexed: 11/25/2022] Open
Abstract
Prostate cancer is the most common non-dermatologic malignancy in men in the Western world. Recently, a frequent chromosomal aberration fusing androgen regulated TMPRSS2 promoter and the ERG gene (TMPRSS2/ERG) was discovered in prostate cancer. Several studies demonstrated cooperation between TMPRSS2/ERG and other defective pathways in cancer progression. However, the unveiling of more specific pathways in which TMPRSS2/ERG takes part, requires further investigation. Using immortalized prostate epithelial cells we were able to show that TMPRSS2/ERG over-expressing cells undergo an Epithelial to Mesenchymal Transition (EMT), manifested by acquisition of mesenchymal morphology and markers as well as migration and invasion capabilities. These findings were corroborated in vivo, where the control cells gave rise to discrete nodules while the TMPRSS2/ERG-expressing cells formed malignant tumors, which expressed EMT markers. To further investigate the general transcription scheme induced by TMPRSS2/ERG, cells were subjected to a microarray analysis that revealed a distinct EMT expression program, including up-regulation of the EMT facilitators, ZEB1 and ZEB2, and down-regulation of the epithelial marker CDH1(E-Cadherin). A chromatin immunoprecipitation assay revealed direct binding of TMPRSS2/ERG to the promoter of ZEB1 but not ZEB2. However, TMPRSS2/ERG was able to bind the promoters of the ZEB2 modulators, IL1R2 and SPINT1. This set of experiments further illuminates the mechanism by which the TMPRSS2/ERG fusion affects prostate cancer progression and might assist in targeting TMPRSS2/ERG and its downstream targets in future drug design efforts.
Collapse
|
36
|
Abstract
The lethal consequences of prostate cancer are related to its metastasis to other organ sites. Epithelial-to-mesenchymal transition (EMT) has received considerable attention as a conceptual paradigm to explain invasive and metastatic behavior during cancer progression. EMT is a normal physiologic process by which cells of epithelial origin convert into cells bearing mesenchymal characteristics. It has been proposed that EMT is co-opted by cancer cells during their metastatic dissemination from a primary organ to secondary sites, but the extent to which this recapitulates physiologic EMT remains uncertain. However, there is ample evidence that EMT-like states occur in, and may contribute to, prostate cancer progression and metastasis, and so has become a very active area of research. Here we review this evidence and explore recent studies that have aimed to better define the role and mechanisms of EMT in prostate cancer. While definitive evidence of something akin to physiologic EMT is still lacking in human prostate cancer, this area of research has nonetheless provided new avenues of investigation into the longstanding puzzles of metastasis, therapeutic resistance, and prognostic biomarkers.
Collapse
|
37
|
Smith CL, Baek ST, Sung CY, Tallquist MD. Epicardial-derived cell epithelial-to-mesenchymal transition and fate specification require PDGF receptor signaling. Circ Res 2011; 108:e15-26. [PMID: 21512159 DOI: 10.1161/circresaha.110.235531] [Citation(s) in RCA: 260] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
RATIONALE In early heart development, platelet-derived growth factor (PDGF) receptor expression in the heart ventricles is restricted to the epicardium. Previously, we showed that PDGFRβ is required for coronary vascular smooth muscle cell (cVSMC) development, but a role for PDGFRα has not been identified. Therefore, we investigated the combined and independent roles of these receptors in epicardial development. OBJECTIVE To understand the contribution of PDGF receptors in epicardial development and epicardial-derived cell fate determination. METHODS AND RESULTS By generating mice with epicardial-specific deletion of the PDGF receptors, we found that epicardial epithelial-to-mesenchymal transition (EMT) was defective. Sox9, an SRY-related transcription factor, was reduced in PDGF receptor-deficient epicardial cells, and overexpression of Sox9 restored epicardial migration, actin reorganization, and EMT gene expression profiles. The failure of epicardial EMT resulted in hearts that lacked epicardial-derived cardiac fibroblasts and cVSMC. Loss of PDGFRα resulted in a specific disruption of cardiac fibroblast development, whereas cVSMC development was unperturbed. CONCLUSIONS Signaling through both PDGF receptors is necessary for epicardial EMT and formation of epicardial-mesenchymal derivatives. PDGF receptors also have independent functions in the development of specific epicardial-derived cell fates.
Collapse
Affiliation(s)
- Christopher L Smith
- Department of Molecular Biology, MC9148, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | | | | | | |
Collapse
|
38
|
Li B, Zheng YW, Sano Y, Taniguchi H. Evidence for mesenchymal-epithelial transition associated with mouse hepatic stem cell differentiation. PLoS One 2011; 6:e17092. [PMID: 21347296 PMCID: PMC3037942 DOI: 10.1371/journal.pone.0017092] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 01/18/2011] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Mesenchymal-epithelial transition events are related to embryonic development, tissue construction, and wound healing. Stem cells are involved in all of these processes, at least in part. However, the direct evidence of mesenchymal-epithelial transition associated with stem cells is unclear. To determine whether mesenchymal-epithelial transition occurs in liver development and/or the differentiation process of hepatic stem cells in vitro, we analyzed a variety of murine liver tissues from embryonic day 11.5 to adults and the colonies derived from hepatic stem/progenitor cells isolated with flow cytometry. The results of gene expression, immunohistochemistry and Western blot showed that as liver develops, the expression of epithelial markers such as Cytokeratin18 and E-cadherin increase, while expression of mesenchymal markers such as vimentin and N-cadherin decreased. On the other hand, in freshly isolated hepatic stem cells, the majority of cells (65.0%) co-express epithelial and mesenchymal markers; this proportion is significantly higher than observed in hematopoietic cells, non-hematopoietic cells and non-stem cell fractions. Likewise, in stem cell-derived colonies cultured over time, upregulation of epithelial genes (Cytokeratin-18 and E-cadherin) occurred simultaneously with downregulation of mesenchymal genes (vimentin and Snail1). Furthermore, in the fetal liver, vimentin-positive cells in the non-hematopoietic fraction had distinct proliferative activity and expressed early the hepatic lineage marker alpha-fetoprotein. CONCLUSION Hepatic stem cells co-express mesenchymal and epithelial markers; the mesenchymal-epithelial transition occurred in both liver development and differentiation of hepatic stem/progenitor cells in vitro. Besides as a mesenchymal marker, vimentin is a novel indicator for cell proliferative activity and undifferentiated status in liver cells.
Collapse
Affiliation(s)
- Bin Li
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yun-Wen Zheng
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuuki Sano
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
39
|
Indra I, Undyala V, Kandow C, Thirumurthi U, Dembo M, Beningo KA. An in vitro correlation of mechanical forces and metastatic capacity. Phys Biol 2011; 8:015015. [PMID: 21301068 DOI: 10.1088/1478-3975/8/1/015015] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mechanical forces have a major influence on cell migration and are predicted to significantly impact cancer metastasis, yet this idea is currently poorly defined. In this study we have asked if changes in traction stress and migratory properties correlate with the metastatic progression of tumor cells. For this purpose, four murine breast cancer cell lines derived from the same primary tumor, but possessing increasing metastatic capacity, were tested for adhesion strength, traction stress, focal adhesion organization and for differential migration rates in two-dimensional and three-dimensional environments. Using traction force microscopy (TFM), we were surprised to find an inverse relationship between traction stress and metastatic capacity, such that force production decreased as the metastatic capacity increased. Consistent with this observation, adhesion strength exhibited an identical profile to the traction data. A count of adhesions indicated a general reduction in the number as metastatic capacity increased but no difference in the maturation as determined by the ratio of nascent to mature adhesions. These changes correlated well with a reduction in active beta-1 integrin with increasing metastatic ability. Finally, in two dimensions, wound healing, migration and persistence were relatively low in the entire panel, maintaining a downward trend with increasing metastatic capacity. Why metastatic cells would migrate so poorly prompted us to ask if the loss of adhesive parameters in the most metastatic cells indicated a switch to a less adhesive mode of migration that would only be detected in a three-dimensional environment. Indeed, in three-dimensional migration assays, the most metastatic cells now showed the greatest linear speed. We conclude that traction stress, adhesion strength and rate of migration do indeed change as tumor cells progress in metastatic capacity and do so in a dimension-sensitive manner.
Collapse
Affiliation(s)
- Indrajyoti Indra
- Department of Biological Science, Wayne State University, Detroit, MI, USA
| | | | | | | | | | | |
Collapse
|
40
|
Patki S, Kadam S, Chandra V, Bhonde R. Human breast milk is a rich source of multipotent mesenchymal stem cells. Hum Cell 2010; 23:35-40. [PMID: 20712706 DOI: 10.1111/j.1749-0774.2010.00083.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Putative stem cells have been isolated from various tissue fluids such as synovial fluid, amniotic fluid, menstrual blood, etc. Recently the presence of nestin positive putative mammary stem cells has been reported in human breast milk. However, it is not clear whether they demonstrate multipotent nature. Since human breast milk is a non-invasive source of mammary stem cells, we were interested in examining the nature of these stem cells. In this pursuit, we could succeed in isolating and expanding a mesenchymal stem cell-like population from human breast milk. These cultured cells were examined by immunofluorescent labeling and found positive for mesenchymal stem cell surface markers CD44, CD29, SCA-1 and negative for CD33, CD34, CD45, CD73 confirming their identity as mesenchymal stem cells. Cytoskeletal protein marker analysis revealed that these cells expressed mesenchymal stem cells markers, namely, nestin, vimentin, smooth muscle actin and also manifests presence of E-Cadherin, an epithelial to mesenchymal transition marker in their early passages. Further we tested the multipotent differentiation potential of these cells and found that they can differentiate into adipogenic, chondrogenic and oesteogenic lineage under the influence of specific differentiation cocktails. This means that these mesenchymal stem cells isolated from human breast milk could potentially be "reprogrammed" to form many types of human tissues. The presence of multipotent stem cells in human milk suggests that breast milk could be an alternative source of stem cells for autologous stem cell therapy although the significance of these cells needs to be determined.
Collapse
Affiliation(s)
- Satish Patki
- Patki Research Foundation and Hospital, Shahupuri, Kolhapur, India
| | | | | | | |
Collapse
|
41
|
Wei W, Barron PD, Rheinwald JG. Modulation of TGF-β-inducible hypermotility by EGF and other factors in human prostate epithelial cells and keratinocytes. In Vitro Cell Dev Biol Anim 2010; 46:841-55. [PMID: 21042878 PMCID: PMC3568941 DOI: 10.1007/s11626-010-9353-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2010] [Accepted: 09/27/2010] [Indexed: 11/28/2022]
Abstract
Keratinocytes migrating from a wound edge or initiating malignant invasion greatly increase their expression of the basement membrane protein Laminin-322 (Lam332). In culture, keratinocytes initiate sustained directional hypermotility when plated onto an incompletely processed form of Lam332 (Lam332') or when treated with transforming growth factor beta (TGF-β), an inducer of Lam332 expression. The development and tissue architecture of stratified squamous and prostate epithelia are very different, yet the basal cells of both express p63, α6β4 integrin, and Lam332. Keratinocytes and prostate epithelial cells grow well in nutritionally optimized culture media with pituitary extract and certain mitogens. We report that prostate epithelial cells display hypermotility responses indistinguishable from those of keratinocytes. Several culture medium variables attenuated TGF-β-induced hypermotility, including Ca(++), serum, and some pituitary extract preparations, without impairing growth, TGF-β growth inhibition, or hypermotility on Lam322'. Distinct from its role as a mitogen, EGF proved to be a required cofactor for TGF-β-induced hypermotility and could not be replaced by HGF or KGF. Prostate epithelial cells have a short replicative lifespan, restricted both by p16(INK4A) and telomere-related mechanisms. We immortalized the normal prostate epithelial cell line HPrE-1 by transduction to express bmi1 and TERT. Prostate epithelial cells lose expression of p63, β4 integrin, and Lam332 when they transform to invasive carcinoma. In contrast, HPrE-1/bmi1/TERT cells retained expression of these proteins and normal TGF-β signaling and hypermotility for >100 doublings. Thus, keratinocytes and prostate epithelial cells possess common hypermotility and senescence mechanisms and immortalized prostate cell lines can be engineered using defined methods to yield cells retaining normal properties.
Collapse
Affiliation(s)
- Wei Wei
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China, 250012
| | - Patricia D. Barron
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - James G. Rheinwald
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
42
|
Ke XS, Qu Y, Cheng Y, Li WC, Rotter V, Øyan AM, Kalland KH. Global profiling of histone and DNA methylation reveals epigenetic-based regulation of gene expression during epithelial to mesenchymal transition in prostate cells. BMC Genomics 2010; 11:669. [PMID: 21108828 PMCID: PMC3012672 DOI: 10.1186/1471-2164-11-669] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 11/25/2010] [Indexed: 12/24/2022] Open
Abstract
Background Previously we reported extensive gene expression reprogramming during epithelial to mesenchymal transition (EMT) of primary prostate cells. Here we investigated the hypothesis that specific histone and DNA methylations are involved in coordination of gene expression during EMT. Results Genome-wide profiling of histone methylations (H3K4me3 and H3K27me3) and DNA methylation (DNAMe) was applied to three cell lines at different stages of a stepwise prostate cell model involving EMT and subsequent accumulation of malignant features. Integrated analyses of epigenetic promoter modifications and gene expression changes revealed strong correlations between the dynamic changes of histone methylations and gene expression. DNA methylation was weaker associated with global gene repression, but strongly correlated to gene silencing when genes co-modified by H3K4me3 were excluded. For genes labeled with multiple epigenetic marks in their promoters, the level of transcription was associated with the net signal intensity of the activating mark H3K4me3 minus the repressive marks H3K27me3 or DNAMe, indicating that the effect on gene expression of bivalent marks (H3K4/K27me3 or H3K4me3/DNAMe) depends on relative modification intensities. Sets of genes, including epithelial cell junction and EMT associated fibroblast growth factor receptor genes, showed corresponding changes concerning epigenetic modifications and gene expression during EMT. Conclusions This work presents the first blueprint of epigenetic modifications in an epithelial cell line and the progeny that underwent EMT and shows that specific histone methylations are extensively involved in gene expression reprogramming during EMT and subsequent accumulation of malignant features. The observation that transcription activity of bivalently marked genes depends on the relative labeling intensity of individual marks provides a new view of quantitative regulation of epigenetic modification.
Collapse
Affiliation(s)
- Xi-Song Ke
- The Gade Institute, University of Bergen, Laboratory Building, 5, etg, Vest, Helse Bergen, NO-5021 Bergen, Norway.
| | | | | | | | | | | | | |
Collapse
|
43
|
Ke XS, Li WC, Hovland R, Qu Y, Liu RH, McCormack E, Thorsen F, Olsen JR, Molven A, Kogan-Sakin I, Rotter V, Akslen LA, Oyan AM, Kalland KH. Reprogramming of cell junction modules during stepwise epithelial to mesenchymal transition and accumulation of malignant features in vitro in a prostate cell model. Exp Cell Res 2010; 317:234-47. [PMID: 20969863 DOI: 10.1016/j.yexcr.2010.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 09/28/2010] [Accepted: 10/09/2010] [Indexed: 12/23/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is pivotal in tumor metastasis. Our previous work reported an EMT model based on primary prostate epithelial cells (EP156T) which gave rise to cells with mesenchymal phenotype (EPT1) without malignant transformation. To promote prostate cell transformation, cells were maintained in saturation density cultures to select for cells overriding quiescence. Foci formed repeatedly following around 8 weeks in confluent EPT1 monolayers. Only later passage EPT1, but not EP156T cells of any passage, could form foci. Cells isolated from the foci were named EPT2 and formed robust colonies in soft agar, a malignant feature present neither in EP156T nor in EPT1 cells. EPT2 cells showed additional malignant traits in vitro, including higher ability to proliferate following confluence, higher resistance to apoptosis and lower dependence on exogenous growth factors than EP156T and EPT1 cells. Microarray profiling identified gene sets, many of which belong to cell junction modules, that changed expression from EP156T to EPT1 cells and continued to change from EPT1 to EPT2 cells. Our findings provide a novel stepwise cell culture model in which EMT emerges independently of transformation and is associated with subsequent accumulation of malignant features in prostate cells. Reprogramming of cell junction modules is involved in both steps.
Collapse
Affiliation(s)
- Xi-song Ke
- The Gade Institute, University of Bergen, Bergen, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kandouz M, Alachkar A, Zhang L, Dekhil H, Chehna F, Yasmeen A, Al Moustafa AE. Teucrium polium plant extract inhibits cell invasion and motility of human prostate cancer cells via the restoration of the E-cadherin/catenin complex. JOURNAL OF ETHNOPHARMACOLOGY 2010; 129:410-415. [PMID: 19897022 DOI: 10.1016/j.jep.2009.10.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2009] [Revised: 09/09/2009] [Accepted: 10/29/2009] [Indexed: 05/28/2023]
Abstract
Prostate cancer is the first most common malignancy in men worldwide; this cancer is characterized by a marked propensity for invasion and spreading to local lymph nodes. On the other hand, Teucrium polium (TP) is a medicinal plant that has been used for more than two thousand years for treating many diseases such as abdominal pain, indigestion and diabetes in the Middle East. However, the effect of TP plant extract on human metastatic cancer cells especially prostate has not been investigated yet. In this study, we examined the effects of TP extract on selected parameters in PC3 and DU145 prostate cancer cell lines. Our results show that TP plant extract inhibits cell proliferation and provokes S cell cycle arrest and reduction of G0-G1 phase. In parallel, this extract induces differentiation to an epithelial phenotype "mesenchymal-epithelial transition" which is an important event in cell invasion and metastasis; thus TP plant extract causes a dramatic decrease in cell invasion and motility abilities of PC3 and DU145 cancer cells in comparison with untreated cells. These changes are accompanied by a re-localization of the expression patterns of E-cadherin and catenins. The molecular pathway analysis of the TP plant extract revealed that it inhibits the phosphorylation of beta-catenin, via Src dephosphorylation, and consequently converts its role from a transcriptional regulator to a cell-cell adhesion molecule. Our findings indicate that TP plant extract inhibits signaling pathways involved in regulating the E-cadherin/catenin complex and possibly other cell-cell adhesion genes via beta-catenin alteration, suggesting that this plant extract has therapeutic promise in the treatment of human metastatic prostate cancer.
Collapse
Affiliation(s)
- Mustapha Kandouz
- Segal Cancer Centre, Lady Davis Institute for Medical Research of the Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Härmä V, Virtanen J, Mäkelä R, Happonen A, Mpindi JP, Knuuttila M, Kohonen P, Lötjönen J, Kallioniemi O, Nees M. A comprehensive panel of three-dimensional models for studies of prostate cancer growth, invasion and drug responses. PLoS One 2010; 5:e10431. [PMID: 20454659 PMCID: PMC2862707 DOI: 10.1371/journal.pone.0010431] [Citation(s) in RCA: 269] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Accepted: 03/31/2010] [Indexed: 01/06/2023] Open
Abstract
Prostate epithelial cells from both normal and cancer tissues, grown in three-dimensional (3D) culture as spheroids, represent promising in vitro models for the study of normal and cancer-relevant patterns of epithelial differentiation. We have developed the most comprehensive panel of miniaturized prostate cell culture models in 3D to date (n = 29), including many non-transformed and most currently available classic prostate cancer (PrCa) cell lines. The purpose of this study was to analyze morphogenetic properties of PrCa models in 3D, to compare phenotypes, gene expression and metabolism between 2D and 3D cultures, and to evaluate their relevance for pre-clinical drug discovery, disease modeling and basic research. Primary and non-transformed prostate epithelial cells, but also several PrCa lines, formed well-differentiated round spheroids. These showed strong cell-cell contacts, epithelial polarization, a hollow lumen and were covered by a complete basal lamina (BL). Most PrCa lines, however, formed large, poorly differentiated spheroids, or aggressively invading structures. In PC-3 and PC-3M cells, well-differentiated spheroids formed, which were then spontaneously transformed into highly invasive cells. These cell lines may have previously undergone an epithelial-to-mesenchymal transition (EMT), which is temporarily suppressed in favor of epithelial maturation by signals from the extracellular matrix (ECM). The induction of lipid and steroid metabolism, epigenetic reprogramming, and ECM remodeling represents a general adaptation to 3D culture, regardless of transformation and phenotype. In contrast, PI3-Kinase, AKT, STAT/interferon and integrin signaling pathways were particularly activated in invasive cells. Specific small molecule inhibitors targeted against PI3-Kinase blocked invasive cell growth more effectively in 3D than in 2D monolayer culture, or the growth of normal cells. Our panel of cell models, spanning a wide spectrum of phenotypic plasticity, supports the investigation of different modes of cell migration and tumor morphologies, and will be useful for predictive testing of anti-cancer and anti-metastatic compounds.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Proliferation/drug effects
- Cell Shape/drug effects
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Collagen/pharmacology
- Drug Combinations
- Epithelial Cells/drug effects
- Epithelial Cells/pathology
- Epithelium/drug effects
- Epithelium/pathology
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Laminin/pharmacology
- Male
- Mesoderm/drug effects
- Mesoderm/pathology
- Models, Biological
- Neoplasm Invasiveness
- Neoplasm Proteins/metabolism
- Phenotype
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Principal Component Analysis
- Prostate/drug effects
- Prostate/pathology
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/enzymology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Proteoglycans/pharmacology
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction/drug effects
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/enzymology
- Spheroids, Cellular/pathology
- TOR Serine-Threonine Kinases
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Ville Härmä
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
| | | | - Rami Mäkelä
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
| | - Antti Happonen
- Knowledge Intensive Services, VTT Technical Research Centre of Finland, Tampere, Finland
| | | | | | - Pekka Kohonen
- Biotechnology Centre, University of Turku, Turku, Finland
| | - Jyrki Lötjönen
- Knowledge Intensive Services, VTT Technical Research Centre of Finland, Tampere, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Matthias Nees
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- * E-mail:
| |
Collapse
|
46
|
Gorlov IP, Gallick GE, Gorlova OY, Amos C, Logothetis CJ. GWAS meets microarray: are the results of genome-wide association studies and gene-expression profiling consistent? Prostate cancer as an example. PLoS One 2009; 4:e6511. [PMID: 19652704 PMCID: PMC2714961 DOI: 10.1371/journal.pone.0006511] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Accepted: 06/29/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) and global profiling of gene expression (microarrays) are two major technological breakthroughs that allow hypothesis-free identification of candidate genes associated with tumorigenesis. It is not obvious whether there is a consistency between the candidate genes identified by GWAS (GWAS genes) and those identified by profiling gene expression (microarray genes). METHODOLOGY/PRINCIPAL FINDINGS We used the Cancer Genetic Markers Susceptibility database to retrieve single nucleotide polymorphisms from candidate genes for prostate cancer. In addition, we conducted a large meta-analysis of gene expression data in normal prostate and prostate tumor tissue. We identified 13,905 genes that were interrogated by both GWASs and microarrays. On the basis of P values from GWASs, we selected 1,649 most significantly associated genes for functional annotation by the Database for Annotation, Visualization and Integrated Discovery. We also conducted functional annotation analysis using same number of the top genes identified in the meta-analysis of the gene expression data. We found that genes involved in cell adhesion were overrepresented among both the GWAS and microarray genes. CONCLUSIONS/SIGNIFICANCE We conclude that the results of these analyses suggest that combining GWAS and microarray data would be a more effective approach than analyzing individual datasets and can help to refine the identification of candidate genes and functions associated with tumor development.
Collapse
Affiliation(s)
- Ivan P Gorlov
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | | |
Collapse
|
47
|
Moen I, Øyan AM, Kalland KH, Tronstad KJ, Akslen LA, Chekenya M, Sakariassen PØ, Reed RK, Stuhr LEB. Hyperoxic treatment induces mesenchymal-to-epithelial transition in a rat adenocarcinoma model. PLoS One 2009; 4:e6381. [PMID: 19636430 PMCID: PMC2712688 DOI: 10.1371/journal.pone.0006381] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Accepted: 06/17/2009] [Indexed: 11/18/2022] Open
Abstract
Tumor hypoxia is relevant for tumor growth, metabolism and epithelial-to-mesenchymal transition (EMT). We report that hyperbaric oxygen (HBO) treatment induced mesenchymal-to-epithelial transition (MET) in a dimetyl-α-benzantracene induced mammary rat adenocarcinoma model, and the MET was associated with extensive coordinated gene expression changes and less aggressive tumors. One group of tumor bearing rats was exposed to HBO (2 bar, pO2 = 2 bar, 4 exposures à 90 minutes), whereas the control group was housed under normal atmosphere (1 bar, pO2 = 0.2 bar). Treatment effects were determined by assessment of tumor growth, tumor vascularisation, tumor cell proliferation, cell death, collagen fibrils and gene expression profile. Tumor growth was significantly reduced (∼16%) after HBO treatment compared to day 1 levels, whereas control tumors increased almost 100% in volume. Significant decreases in tumor cell proliferation, tumor blood vessels and collagen fibrils, together with an increase in cell death, are consistent with tumor growth reduction and tumor stroma influence after hyperoxic treatment. Gene expression profiling showed that HBO induced MET. In conclusion, hyperoxia induced MET with coordinated expression of gene modules involved in cell junctions and attachments together with a shift towards non-tumorigenic metabolism. This leads to more differentiated and less aggressive tumors, and indicates that oxygen per se might be an important factor in the “switches” of EMT and MET in vivo. HBO treatment also attenuated tumor growth and changed tumor stroma, by targeting the vascular system, having anti-proliferative and pro-apoptotic effects.
Collapse
Affiliation(s)
- Ingrid Moen
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Anne Margrete Øyan
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Karl-Henning Kalland
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | | | - Lars Andreas Akslen
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Martha Chekenya
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Rolf Kåre Reed
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | |
Collapse
|
48
|
Abstract
Viruses enter host cells in order to complete their life cycles and have evolved to exploit host cell structures, regulatory factors and mechanisms. The virus and host cell interactions have consequences at multiple levels, spanning from evolution through disease to models and tools for scientific discovery and treatment. Virus-induced human cancers arise after a long duration of time and are monoclonal or oligoclonal in origin. Cancer is therefore a side effect rather than an essential part of viral infections in humans. Still, 15-20% of all human cancers are caused by viruses. A review of tumour virology shows its close integration in cancer research. Viral tools and experimental models have been indispensible for the progress of molecular biology. In particular, retroviruses and DNA tumour viruses have played major roles in our present understanding of the molecular biology of both viruses and the host. Recently, additional complex relationships due to virus and host co-evolution have appeared and may lead to a further understanding of the overall regulation of gene expression programmes in cancer.
Collapse
|
49
|
Ke XS, Qu Y, Rostad K, Li WC, Lin B, Halvorsen OJ, Haukaas SA, Jonassen I, Petersen K, Goldfinger N, Rotter V, Akslen LA, Oyan AM, Kalland KH. Genome-wide profiling of histone h3 lysine 4 and lysine 27 trimethylation reveals an epigenetic signature in prostate carcinogenesis. PLoS One 2009; 4:e4687. [PMID: 19262738 PMCID: PMC2650415 DOI: 10.1371/journal.pone.0004687] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Accepted: 12/25/2008] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Increasing evidence implicates the critical roles of epigenetic regulation in cancer. Very recent reports indicate that global gene silencing in cancer is associated with specific epigenetic modifications. However, the relationship between epigenetic switches and more dynamic patterns of gene activation and repression has remained largely unknown. METHODOLOGY/PRINCIPAL FINDINGS Genome-wide profiling of the trimethylation of histone H3 lysine 4 (H3K4me3) and lysine 27 (H3K27me3) was performed using chromatin immunoprecipitation coupled with whole genome promoter microarray (ChIP-chip) techniques. Comparison of the ChIP-chip data and microarray gene expression data revealed that loss and/or gain of H3K4me3 and/or H3K27me3 were strongly associated with differential gene expression, including microRNA expression, between prostate cancer and primary cells. The most common switches were gain or loss of H3K27me3 coupled with low effect on gene expression. The least prevalent switches were between H3K4me3 and H3K27me3 coupled with much higher fractions of activated and silenced genes. Promoter patterns of H3K4me3 and H3K27me3 corresponded strongly with coordinated expression changes of regulatory gene modules, such as HOX and microRNA genes, and structural gene modules, such as desmosome and gap junction genes. A number of epigenetically switched oncogenes and tumor suppressor genes were found overexpressed and underexpressed accordingly in prostate cancer cells. CONCLUSIONS/SIGNIFICANCE This work offers a dynamic picture of epigenetic switches in carcinogenesis and contributes to an overall understanding of coordinated regulation of gene expression in cancer. Our data indicate an H3K4me3/H3K27me3 epigenetic signature of prostate carcinogenesis.
Collapse
Affiliation(s)
- Xi-Song Ke
- The Gade Institute, University of Bergen, Bergen, Norway
- * E-mail: (X-SK); (K-HK)
| | - Yi Qu
- The Gade Institute, University of Bergen, Bergen, Norway
| | - Kari Rostad
- The Gade Institute, University of Bergen, Bergen, Norway
| | - Wen-Cheng Li
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Biaoyang Lin
- Swedish Medical Center, Seattle, Washington, United States of America
- Zhejiang-California International Nanosystems Institute (ZCNI), Zhejiang University, Hangzhou, People's Republic of China
| | | | - Svein A. Haukaas
- Department of Surgical Sciences, University of Bergen, Bergen, Norway
- Department of Surgery, Haukeland University Hospital, Bergen, Norway
| | - Inge Jonassen
- Computational Biology Unit, Bergen Center for Computational Science, University of Bergen, Bergen, Norway
- Department of Informatics, University of Bergen, Bergen, Norway
| | - Kjell Petersen
- Computational Biology Unit, Bergen Center for Computational Science, University of Bergen, Bergen, Norway
- Department of Informatics, University of Bergen, Bergen, Norway
| | - Naomi Goldfinger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Lars A. Akslen
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Anne M. Oyan
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Microbiology and Immunology, Haukeland University Hospital, Bergen, Norway
| | - Karl-Henning Kalland
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Microbiology and Immunology, Haukeland University Hospital, Bergen, Norway
- * E-mail: (X-SK); (K-HK)
| |
Collapse
|