1
|
Yamamoto T, Sato Y, Yasuda S, Shikamura M, Tamura T, Takenaka C, Takasu N, Nomura M, Dohi H, Takahashi M, Mandai M, Kanemura Y, Nakamura M, Okano H, Kawamata S. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:527-538. [PMID: 35445254 PMCID: PMC9154342 DOI: 10.1093/stcltm/szac014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/13/2022] [Indexed: 11/15/2022] Open
Abstract
Cell therapy using induced pluripotent stem cell (iPSC) derivatives may result in abnormal tissue generation because the cells undergo numerous cycles of mitosis before clinical application, potentially increasing the accumulation of genetic abnormalities. Therefore, genetic tests may predict abnormal tissue formation after transplantation. Here, we administered iPSC derivatives with or without single-nucleotide variants (SNVs) and deletions in cancer-related genes with various genomic copy number variant (CNV) profiles into immunodeficient mice and examined the relationships between mutations and abnormal tissue formation after transplantation. No positive correlations were found between the presence of SNVs/deletions and the formation of abnormal tissues; the overall predictivity was 29%. However, a copy number higher than 3 was correlated, with an overall predictivity of 86%. Furthermore, we found CNV hotspots at 14q32.33 and 17q12 loci. Thus, CNV analysis may predict abnormal tissue formation after transplantation of iPSC derivatives and reduce the number of tumorigenicity tests.
Collapse
Affiliation(s)
- Takako Yamamoto
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Masayuki Shikamura
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Takashi Tamura
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Chiemi Takenaka
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | | | | | | | | | | | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Shin Kawamata
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
- Riken BDR, Kobe, Japan
- Corresponding author: Shin Kawamata, Minatojima-minamimachi 1-5-4, Chuo-ku Kobe, 650-0047 Japan.
| |
Collapse
|
2
|
Evaluation of two in vitro assays for tumorigenicity assessment of CRISPR-Cas9 genome-edited cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:241-253. [PMID: 34703845 PMCID: PMC8505356 DOI: 10.1016/j.omtm.2021.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/03/2021] [Indexed: 12/26/2022]
Abstract
Off-target editing is one of the main safety concerns for the use of CRISPR-Cas9 genome editing in gene therapy. These unwanted modifications could lead to malignant transformation, which renders tumorigenicity assessment of gene therapy products indispensable. In this study, we established two in vitro transformation assays, the soft agar colony-forming assay (SACF) and the growth in low attachment assay (GILA) as alternative methods for tumorigenicity evaluation of genome-edited cells. Using a CRISPR-Cas9-based approach to transform immortalized MCF10A cells, we identified PTPN12, a known tumor suppressor, as a valid positive control in GILA and SACF. Next, we measured the limit of detection for both assays and proved that SACF is more sensitive than GILA (0.8% versus 3.1% transformed cells). We further validated SACF and GILA by identifying a set of positive and negative controls and by testing the suitability of another cell line (THLE-2). Moreover, in contrast to SACF and GILA, an in vivo tumorigenicity study failed to detect the known tumorigenic potential of PTPN12 deletion, demonstrating the relevance of GILA and SACF in tumorigenicity testing. In conclusion, SACF and GILA are both attractive and valuable additions to preclinical safety assessment of gene therapy products.
Collapse
|
3
|
|
4
|
Inhibitors of DNA double-strand break repair at the crossroads of cancer therapy and genome editing. Biochem Pharmacol 2020; 182:114195. [DOI: 10.1016/j.bcp.2020.114195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/03/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022]
|
5
|
Hu W, Li S, Zhang S, Xie B, Zheng M, Sun J, Yang X, Zang L. GJA1 is a Prognostic Biomarker and Correlated with Immune Infiltrates in Colorectal Cancer. Cancer Manag Res 2020; 12:11649-11661. [PMID: 33235496 PMCID: PMC7678473 DOI: 10.2147/cmar.s235500] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 09/24/2020] [Indexed: 12/26/2022] Open
Abstract
Purpose Previous studies found that gap junction alpha-1 protein (GJA1) was a potent tumor suppressor in colorectal cancer (CRC). We designed the present study to evaluate the clinical importance and molecular mechanisms of GJA1 in CRC. Methods Clinical and transcriptomic data from TCGA and GEO datasets were retrospectively collected. CRC patients were divided into two subgroups according to the expression level of GJA1 mRNA. Difference between survival time and response to neoadjuvant chemotherapy was then evaluated. Functional assays including wound-healing assay, transwell invasion assay and flow cytometry assay were performed to investigate the effects of GJA1 on invasive ability and response to chemotherapy drugs of CRC cells. Moreover, we explored the mechanisms of GJA1 by which it regulates CRC malignant phenotypes. Results The expression level of GJA1 was significantly higher in normal tissue than cancer tissue, indicating a tumor suppressive role of GJA1 in CRC. Patients with higher expression of GJA1 showed better prognosis than those with low GJA1 expression level. Consistently, overexpression of GJA1 suppressed the invasive ability of CRC cells while enhancing the sensitivity of CRC cells to oxaliplatin-induced apoptosis. Mechanically, we found that GJA1 suppressed the epithelial mesenchymal transition process. Moreover, GJA1 could modulating infiltrating levels of several immune cells in the tumor microenvironment. Conclusion These findings suggested that GJA1 was correlated with prognosis and immune infiltrating levels of CD8+ T cells, macrophages, neutrophils, and DCs in CRC. In addition, GJA1 expression contributes to regulation of tumor-associated macrophages (TAMs) and tumor infiltrating neutrophils (TINs) in CRC. These findings suggest that GJA1 is a promising biomarker for determining prognosis and immune infiltration in colorectal cancer.
Collapse
Affiliation(s)
- Wenjun Hu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Shuchun Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Sen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bowen Xie
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiao Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Lu Zang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Han W, Wu Q, Zhang X, Duan Z. Innovation for hepatotoxicity in vitro research models: A review. J Appl Toxicol 2018; 39:146-162. [PMID: 30182494 DOI: 10.1002/jat.3711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022]
Abstract
Many categories of drugs can induce hepatotoxicity, so improving the prediction of toxic drugs is important. In vitro models using human hepatocytes are more accurate than in vivo animal models. Good in vitro models require an abundance of metabolic enzyme activities and normal cellular polarity. However, none of the in vitro models can completely simulate hepatocytes in the human body. There are two ways to overcome this limitation: enhancing the metabolic function of hepatocytes and changing the cultural environment. In this review, we summarize the current state of research, including the main characteristics of in vitro models and their limitations, as well as improved technology and developmental prospects. We hope that this review provides some new ideas for hepatotoxicity research.
Collapse
Affiliation(s)
- Weijia Han
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Qiao Wu
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Xiaohui Zhang
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Zhongping Duan
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| |
Collapse
|
7
|
Khatami F, Tavangar SM. Genetic and Epigenetic of Medullary Thyroid Cancer. IRANIAN BIOMEDICAL JOURNAL 2018; 22:142-50. [PMID: 29126344 PMCID: PMC5889499 DOI: 10.22034/ibj.22.3.142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/25/2017] [Accepted: 10/28/2017] [Indexed: 02/06/2023]
Abstract
Medullary thyroid carcinoma (MTC) is an infrequent, calcitonin producing neuroendocrine tumor and initiates from the parafollicular C cells of the thyroid gland. Several genetic and epigenetic alterations are collaterally responsible for medullary thyroid carcinogenesis. In this review article, we shed light on all the genetic and epigenetic hallmarks of MTC. From the genetic perspective, RET, HRAS, and KRAS are the most important genes that are characterized in MTC. From the epigenetic perspective, Ras-association domain family member 1A, telomerase reverse transcriptase promoter methylations, overexpression of histone methyltransferases, EZH2 and SMYD3, and wide ranging increase and decrease in non-coding RNAs can be responsible for medullary thyroid carcinogenesis.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Avivar-Valderas A, McEwen R, Taheri-Ghahfarokhi A, Carnevalli LS, Hardaker EL, Maresca M, Hudson K, Harrington EA, Cruzalegui F. Functional significance of co-occurring mutations in PIK3CA and MAP3K1 in breast cancer. Oncotarget 2018; 9:21444-21458. [PMID: 29765551 PMCID: PMC5940413 DOI: 10.18632/oncotarget.25118] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/22/2018] [Indexed: 12/30/2022] Open
Abstract
The PI3Kα signaling pathway is frequently hyper-activated in breast cancer (BrCa), as a result of mutations/amplifications in oncogenes (e.g. HER2), decreased function in tumor suppressors (e.g. PTEN) or activating mutations in key components of the pathway. In particular, activating mutations of PIK3CA (~45%) are frequently found in luminal A BrCa samples. Genomic studies have uncovered inactivating mutations in MAP3K1 (13-20%) and MAP2K4 (~8%), two upstream kinases of the JNK apoptotic pathway in luminal A BrCa samples. Further, simultaneous mutation of PIK3CA and MAP3K1 are found in ~11% of mutant PIK3CA tumors. How these two alterations may cooperate to elicit tumorigenesis and impact the sensitivity to PI3K and AKT inhibitors is currently unknown. Using CRISPR gene editing we have genetically disrupted MAP3K1 expression in mutant PIK3CA cell lines to specifically create in vitro models reflecting the mutational status of PIK3CA and MAP3K1 in BrCa patients. MAP3K1 deficient cell lines exhibited ~2.4-fold increased proliferation rate and decreased sensitivity to PI3Kα/δ(AZD8835) and AKT (AZD5363) inhibitors (~2.61 and ~5.23-fold IC50 increases, respectively) compared with parental control cell lines. In addition, mechanistic analysis revealed that MAP3K1 disruption enhances AKT phosphorylation and downstream signaling and reduces sensitivity to AZD5363-mediated pathway inhibition. This appears to be a consequence of deficient MAP3K1-JNK signaling increasing IRS1 stability and therefore promoting IRS1 binding to p85, resulting in enhanced PI3Kα activity. Using 3D-MCF10A-PI3KαH1047R models, we found that MAP3K1 depletion increased overall acinar volume and counteracted AZD5363-mediated reduction of acinar growth due to enhanced proliferation and reduced apoptosis. Furthermore, in vivo efficacy studies revealed that MAP3K1-deficient MCF7 tumors were less sensitive to AKT inhibitor treatment, compared with parental MCF7 tumors. Our study provides mechanistic and in vivo evidence indicating a role for MAP3K1 as a tumor suppressor gene at least in the context of PIK3CA-mutant backgrounds. Further, our work predicts that MAP3K1 mutational status may be considered as a predictive biomarker for efficacy in PI3K pathway inhibitor trials.
Collapse
Affiliation(s)
- Alvaro Avivar-Valderas
- Translational Science, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK.,Current address: TiGenix, Parque Tecnológico de Madrid, Tres Cantos, Madrid, Spain
| | - Robert McEwen
- Translational Science, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Amir Taheri-Ghahfarokhi
- Translational Genomics, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | | | | - Marcello Maresca
- Translational Genomics, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Kevin Hudson
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK.,Current address: 2TheNth, Adelphi Mill, Bollington, Macclesfield, UK
| | | | - Francisco Cruzalegui
- Translational Science, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK.,Current address: Pierre Fabre R&D Centre, Toulouse, France
| |
Collapse
|
9
|
Aleskandarany MA, Vandenberghe ME, Marchiò C, Ellis IO, Sapino A, Rakha EA. Tumour Heterogeneity of Breast Cancer: From Morphology to Personalised Medicine. Pathobiology 2018; 85:23-34. [DOI: 10.1159/000477851] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/30/2017] [Indexed: 12/11/2022] Open
|
10
|
Yousafzai MS, Coceano G, Bonin S, Niemela J, Scoles G, Cojoc D. Investigating the effect of cell substrate on cancer cell stiffness by optical tweezers. J Biomech 2017; 60:266-269. [PMID: 28712542 DOI: 10.1016/j.jbiomech.2017.06.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 06/20/2017] [Accepted: 06/29/2017] [Indexed: 01/22/2023]
Abstract
The mechanical properties of cells are influenced by their microenvironment. Here we report cell stiffness alteration by changing the cell substrate stiffness for isolated cells and cells in contact with other cells. Polydimethylsiloxane (PDMS) is used to prepare soft substrates with three different stiffness values (173, 88 and 17kPa respectively). Breast cancer cells lines, namely HBL-100, MCF-7 and MDA-MB-231 with different level of aggressiveness are cultured on these substrates and their local elasticity is investigated by vertical indentation of the cell membrane. Our preliminary results show an unforeseen behavior of the MDA-MB-231 cells. When cultured on glass substrate as isolated cells, they are less stiff than the other two types of cells, in agreement with the general statement that more aggressive and metastatic cells are softer. However, when connected to other cells the stiffness of MDA-MB-231 cells becomes similar to the other two cell lines. Moreover, the stiffness of MDA-MB-231 cells cultured on soft PDMS substrates is significantly higher than the stiffness of the other cell types, demonstrating thus the strong influence of the environmental conditions on the mechanical properties of the cells.
Collapse
Affiliation(s)
- Muhammad Sulaiman Yousafzai
- Institute of Materials (IOM-CNR), Area Science Park, Basovizza, S.S. 14, Km 163.5, 34149 Trieste, Italy; The Abdus Salam International Centre for Theoretical Physics (ICTP), 34149 Trieste, Italy
| | - Giovanna Coceano
- Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Serena Bonin
- Department of Medical Sciences (DSM), University of Trieste, 34149, Italy
| | - Joseph Niemela
- The Abdus Salam International Centre for Theoretical Physics (ICTP), 34149 Trieste, Italy
| | - Giacinto Scoles
- Department of Biological and Medical Science, University of Udine, Santa Maria della Misericordia Hospital, 33100 Udine, Italy
| | - Dan Cojoc
- Institute of Materials (IOM-CNR), Area Science Park, Basovizza, S.S. 14, Km 163.5, 34149 Trieste, Italy.
| |
Collapse
|
11
|
Single cell time-lapse analysis reveals that podoplanin enhances cell survival and colony formation capacity of squamous cell carcinoma cells. Sci Rep 2017; 7:39971. [PMID: 28059107 PMCID: PMC5216406 DOI: 10.1038/srep39971] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/30/2016] [Indexed: 01/04/2023] Open
Abstract
Tumor initiating cells (TICs) are characterized by high clonal expansion capacity. We previously reported that podoplanin is a TIC-specific marker for the human squamous cell carcinoma cell line A431. The aim of this study is to explore the molecular mechanism underlying the high clonal expansion potential of podoplanin-positive A431cells using Fucci imaging. Single podoplanin-positive cells created large colonies at a significantly higher frequency than single podoplanin-negative cells, whereas no difference was observed between the two types of cells with respect to cell cycle status. Conversely, the cell death ratio of progenies derived from podoplanin-positive single cell was significantly lower than that of cells derived from podoplanin-negative cells. Single A431 cells, whose podoplanin expression was suppressed by RNA interference, exhibited increased cell death ratios and decreased frequency of large colony forming. Moreover, the frequency of large colony forming decreased significantly when podoplanin-positive single cells was treated with a ROCK (Rho-associated coiled-coil kinase) inhibitor, whereas no difference was observed in single podoplanin-negative cells. Our current study cleared that high clonal expansion capacity of podoplanin-positive TICs populations was the result of reduced cell death by podoplanin-mediated signaling. Therefore, podoplanin activity may be a therapeutic target in the treatment of squamous cell carcinomas.
Collapse
|
12
|
Parisi L, Zomer Volpato F, Cagol N, Siciliano M, Migliaresi C, Motta A, Sala R. An innovative protocol for schwann cells extracellular matrix proteins extraction. J Biomed Mater Res A 2016; 104:3175-3180. [PMID: 27500379 DOI: 10.1002/jbm.a.35854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 07/15/2016] [Accepted: 08/02/2016] [Indexed: 01/01/2023]
Abstract
The evidence that extracellular matrix (ECM) components could represent new targets for drugs designed to approach degenerative disease, requires their analysis. Before the analysis, proteins should be extracted from ECM and solubilized. Currently, few protocols for ECM proteins extraction and solubilization are available in literature, and most of them are based mainly on the use of proteolytic enzymes, such as trypsin, which often lead to proteins damage. Moreover, no methods have been so far proposed to solubilize Schwann Cell ECM, which may represent an important target for the therapy of neurodegenerative disorders. In our study, we propose to solubilize SC ECM through the use of surfactants and urea. We compared our method of solubilization, with one of that proposed in literature for a general ECM, mainly based on the use of enzymes. We want to highlight the benefit of solubilizing SC ECM, avoiding the use of proteolytic enzymes. To compare the amount of proteins extracted with both methods, MicroBCA assay was used, while the quality of the proteins extracted was observed through the SDS-PAGE. The results obtained confirm a better solubilization of SC ECM proteins with the proposed protocol, both quantitatively and qualitatively, showing a higher concentration of proteins extracted and a better enrichment of protein fractions, if compared to the enzyme-based protocol. Our results show that SC ECM could be efficiently solubilized through the use of surfactant and urea, avoiding the use of enzyme-base methods. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 3175-3180, 2016.
Collapse
Affiliation(s)
- L Parisi
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy.
| | - F Zomer Volpato
- Department of Industrial Engineering, University of Trento, Trento, Italy
| | - N Cagol
- Department of Industrial Engineering, University of Trento, Trento, Italy
| | - M Siciliano
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| | - C Migliaresi
- Department of Industrial Engineering, University of Trento, Trento, Italy
| | - A Motta
- Department of Industrial Engineering, University of Trento, Trento, Italy
| | - R Sala
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| |
Collapse
|
13
|
Abstract
Breast cell pathology results from biochemical and molecular changes that culminate in the cell’s loss of functional responsiveness. The epithelial cell compartment in the breast ductal system is the site of approximately 98% of malignant proliferations, and it is from within these cells that the first biochemical signal of change may be expressed as an inflammatory response. Inflammation may be represented by biomarkers of early pathologic changes in breast cells and be associated with risk for the development of breast cancer. A theoretical model of the inflammatory process is proposed showing predictive linkages among stimuli in the breast microenvironment and the development of breast pathology, in particular, breast cancer. This model fuels intervention concepts that may prevent malignant breast health outcomes.
Collapse
|
14
|
Dick JM. Proteomic indicators of oxidation and hydration state in colorectal cancer. PeerJ 2016; 4:e2238. [PMID: 27547546 PMCID: PMC4958012 DOI: 10.7717/peerj.2238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/20/2016] [Indexed: 12/15/2022] Open
Abstract
New integrative approaches are needed to harness the potential of rapidly growing datasets of protein expression and microbial community composition in colorectal cancer. Chemical and thermodynamic models offer theoretical tools to describe populations of biomacromolecules and their relative potential for formation in different microenvironmental conditions. The average oxidation state of carbon (ZC) can be calculated as an elemental ratio from the chemical formulas of proteins, and water demand per residue (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}${\overline{n}}_{{\mathrm{H}}_{2}\mathrm{O}}$\end{document}n¯H2O) is computed by writing the overall formation reactions of proteins from basis species. Using results reported in proteomic studies of clinical samples, many datasets exhibit higher mean ZC or \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}${\overline{n}}_{{\mathrm{H}}_{2}\mathrm{O}}$\end{document}n¯H2O of proteins in carcinoma or adenoma compared to normal tissue. In contrast, average protein compositions in bacterial genomes often have lower ZC for bacteria enriched in fecal samples from cancer patients compared to healthy donors. In thermodynamic calculations, the potential for formation of the cancer-related proteins is energetically favored by changes in the chemical activity of H2O and fugacity of O2 that reflect the compositional differences. The compositional analysis suggests that a systematic change in chemical composition is an essential feature of cancer proteomes, and the thermodynamic descriptions show that the observed proteomic transformations in host tissue could be promoted by relatively high microenvironmental oxidation and hydration states.
Collapse
|
15
|
Yousafzai MS, Coceano G, Mariutti A, Ndoye F, Amin L, Niemela J, Bonin S, Scoles G, Cojoc D. Effect of neighboring cells on cell stiffness measured by optical tweezers indentation. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:57004. [PMID: 27232596 DOI: 10.1117/1.jbo.21.5.057004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/25/2016] [Indexed: 06/05/2023]
Abstract
We report on the modification of mechanical properties of breast cancer cells when they get in contact with other neighboring cells of the same type. Optical tweezers vertical indentation was employed to investigate cell mechanics in isolated and contact conditions, by setting up stiffness as a marker. Two human breast cancer cell lines with different aggressiveness [MCF-7 (luminal breast cancer) and MDA-MB-231 (basal-like breast cancer)] and one normal immortalized breast cell line HBL-100 (normal and myoepithelial) were selected. We found that neighboring cells significantly alter cell stiffness: MDA-MB-231 becomes stiffer when in contact, while HBL-100 and MCF-7 exhibit softer character. Cell stiffness was probed at three cellular subregions: central (above nucleus), intermediate (cytoplasm), and near the leading edge. In an isolated condition, all cells showed a significant regional variation in stiffness: higher at the center and fading toward the leading edge. However, the regional variation becomes statistically insignificant when the cells were in contact with other neighboring cells. The proposed approach will contribute to understand the intriguing temporal sequential alterations in cancer cells during interaction with their surrounding microenvironment.
Collapse
Affiliation(s)
- Muhammad S Yousafzai
- Institute of Materials (IOM-CNR), Area Science Park, Basovizza, S.S. 14, Km 163.5, 34149 Trieste, ItalybUniversity of Trieste, PhD School of Nanotechnology, Via Alfonso Valerio 2, Trieste 34127, ItalycUniversity of Udine, Department of Biological and Medi
| | - Giovanna Coceano
- Institute of Materials (IOM-CNR), Area Science Park, Basovizza, S.S. 14, Km 163.5, 34149 Trieste, ItalybUniversity of Trieste, PhD School of Nanotechnology, Via Alfonso Valerio 2, Trieste 34127, Italy
| | - Alberto Mariutti
- University of Udine, Department of Biological and Medical Science, Santa Maria della Misericordia Hospital, Piazzale Kolbe 4, Udine 33100, Italy
| | - Fatou Ndoye
- The Abdus Salam International Centre for Theoretical Physics (ICTP), Via Beirut 6, Trieste 34149, ItalyeCheikh Anta Diop University, Department of Physics, Dakar 5005, Senegal
| | - Ladan Amin
- International School for Advanced Studies (SISSA), Neuroscience Area, Via Bonomea 265, Trieste 34136, Italy
| | - Joseph Niemela
- The Abdus Salam International Centre for Theoretical Physics (ICTP), Via Beirut 6, Trieste 34149, Italy
| | - Serena Bonin
- University of Trieste, Department of Medical Sciences, Cattinara Hospital, Strada di Fiume 447, Trieste 34149, Italy
| | - Giacinto Scoles
- University of Udine, Department of Biological and Medical Science, Santa Maria della Misericordia Hospital, Piazzale Kolbe 4, Udine 33100, Italy
| | - Dan Cojoc
- Institute of Materials (IOM-CNR), Area Science Park, Basovizza, S.S. 14, Km 163.5, 34149 Trieste, Italy
| |
Collapse
|
16
|
Ruszczyk M, Zirpoli G, Kumar S, Bandera EV, Bovbjerg DH, Jandorf L, Khoury T, Hwang H, Ciupak G, Pawlish K, Schedin P, Masso-Welch P, Ambrosone CB, Hong CC. Breast cancer risk factor associations differ for pure versus invasive carcinoma with an in situ component in case-control and case-case analyses. Cancer Causes Control 2015; 27:183-98. [PMID: 26621543 DOI: 10.1007/s10552-015-0696-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 11/07/2015] [Indexed: 12/29/2022]
Abstract
PURPOSE Invasive ductal carcinoma (IDC) is diagnosed with or without a ductal carcinoma in situ (DCIS) component. Previous analyses have found significant differences in tumor characteristics between pure IDC lacking DCIS and mixed IDC with DCIS. We will test our hypothesis that pure IDC represents a form of breast cancer with etiology and risk factors distinct from mixed IDC/DCIS. METHODS We compared reproductive risk factors for breast cancer risk, as well as family and smoking history between 831 women with mixed IDC/DCIS (n = 650) or pure IDC (n = 181), and 1,620 controls, in the context of the Women's Circle of Health Study (WCHS), a case-control study of breast cancer in African-American and European-American women. Data on reproductive and lifestyle factors were collected during interviews, and tumor characteristics were abstracted from pathology reports. Case-control and case-case analyses were conducted using unconditional logistic regression. RESULTS Most risk factors were similarly associated with pure IDC and mixed IDC/DCIS. However, among postmenopausal women, risk of pure IDC was lower in women with body mass index (BMI) 25 to <30 [odds ratio (OR) 0.66; 95 % confidence interval (CI) 0.35-1.23] and BMI ≥ 30 (OR 0.33; 95 % CI 0.18-0.67) compared to women with BMI < 25, with no associations with mixed IDC/DCIS. In case-case analyses, women who breastfed up to 12 months (OR 0.55; 95 % CI 0.32-0.94) or longer (OR 0.47; 95 % CI 0.26-0.87) showed decreased odds of pure IDC than mixed IDC/DCIS compared to those who did not breastfeed. CONCLUSIONS Associations with some breast cancer risk factors differed between mixed IDC/DCIS and pure IDC, potentially suggesting differential developmental pathways. These findings, if confirmed in a larger study, will provide a better understanding of the developmental patterns of breast cancer and the influence of modifiable risk factors, which in turn could lead to better preventive measures for pure IDC, which have worse disease prognosis compared to mixed IDC/DCIS.
Collapse
Affiliation(s)
- Melanie Ruszczyk
- Department of Biotechnical and Clinical Laboratory Sciences, University at Buffalo, 12 Capen Hall, Buffalo, NY, 14214, USA.
| | - Gary Zirpoli
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm & Carlton Sts., Buffalo, NY, 14263, USA.
| | - Shicha Kumar
- Department of Surgical Oncology, Roswell Park Cancer Institute, Elm & Carlton Sts., Buffalo, NY, 14263, USA.
| | - Elisa V Bandera
- Population Science/Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, 195 Little Albany St., New Brunswick, NJ, 08903, USA. .,Department of Epidemiology, Rutgers School of Public Health, 683 Hoes Ln W, Piscataway, NJ, 08854, USA.
| | - Dana H Bovbjerg
- Department of Psychiatry, University of Pittsburgh Cancer Institute, 5150 Centre Ave., Pittsburgh, PA, 15232, USA.
| | - Lina Jandorf
- Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, 1428 Madison Ave., New York, NY, 10029, USA.
| | - Thaer Khoury
- Department of Pathology, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY, 14263, USA.
| | - Helena Hwang
- Department of Pathology, University of Texas, Southwestern Medical Center, 5325 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Gregory Ciupak
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm & Carlton Sts., Buffalo, NY, 14263, USA.
| | - Karen Pawlish
- New Jersey State Cancer Registry, New Jersey Department of Health, 140 East Front Street, Trenton, NJ, 08625, USA.
| | - Pepper Schedin
- Department of Cell, Development and Cancer Biology, Oregon Health Sciences University, 3181 SW Sam Jackson Pkwy, Portland, OR, 97239, USA.
| | - Patricia Masso-Welch
- Department of Biotechnical and Clinical Laboratory Sciences, University at Buffalo, 12 Capen Hall, Buffalo, NY, 14214, USA.
| | - Christine B Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm & Carlton Sts., Buffalo, NY, 14263, USA.
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm & Carlton Sts., Buffalo, NY, 14263, USA.
| |
Collapse
|
17
|
Walker ND, Patel J, Munoz JL, Hu M, Guiro K, Sinha G, Rameshwar P. The bone marrow niche in support of breast cancer dormancy. Cancer Lett 2015; 380:263-71. [PMID: 26546045 DOI: 10.1016/j.canlet.2015.10.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/13/2015] [Accepted: 10/27/2015] [Indexed: 12/15/2022]
Abstract
Despite the success in detecting breast cancer (BC) early and, with aggressive therapeutic intervention, BC remains a clinical problem. The bone marrow (BM) is a favorable metastatic site for breast cancer cells (BCCs). In BM, the survival of BCCs is partly achieved by the supporting microenvironment, including the presence of immune suppressive cells such as mesenchymal stem cells (MSCs). The heterogeneity of BCCs brings up the question of how each subset interacts with the BM microenvironment. The cancer stem cells (CSCs) survive in the BM as cycling quiescence cells and, forming gap junctional intercellular communication (GJIC) with the hematopoietic supporting stromal cells and MSCs. This type of communication has been identified close to the endosteum. Additionally, dormancy can occur by soluble mediators such as cytokines and also by the exchange of exosomes. These latter mechanisms are reviewed in the context of metastasis of BC to the BM for transition as dormant cells. The article also discusses how immune cells such as macrophages and regulatory T-cells facilitate BC dormancy. The challenges of studying BC dormancy in 2-dimensional (2-D) system are also incorporated by proposing 3-D system by engineering methods to recapitulate the BM microenvironment.
Collapse
Affiliation(s)
- Nykia D Walker
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA; Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Jimmy Patel
- Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Jessian L Munoz
- Ob/Gyn and Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Madeleine Hu
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA; Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Khadidiatou Guiro
- Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Garima Sinha
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA; Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA; Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
18
|
White MJV, Roife D, Gomer RH. Galectin-3 Binding Protein Secreted by Breast Cancer Cells Inhibits Monocyte-Derived Fibrocyte Differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:1858-67. [PMID: 26136428 PMCID: PMC4530092 DOI: 10.4049/jimmunol.1500365] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/02/2015] [Indexed: 12/26/2022]
Abstract
To metastasize, tumor cells often need to migrate through a layer of collagen-containing scar tissue which encapsulates the tumor. A key component of scar tissue and fibrosing diseases is the monocyte-derived fibrocyte, a collagen-secreting profibrotic cell. To test the hypothesis that invasive tumor cells may block the formation of the fibrous sheath, we determined whether tumor cells secrete factors that inhibit monocyte-derived fibrocyte differentiation. We found that the human metastatic breast cancer cell line MDA-MB-231 secretes activity that inhibits human monocyte-derived fibrocyte differentiation, whereas less aggressive breast cancer cell lines secrete less of this activity. Purification indicated that Galectin-3 binding protein (LGALS3BP) is the active factor. Recombinant LGALS3BP inhibits monocyte-derived fibrocyte differentiation, and immunodepletion of LGALS3BP from MDA-MB 231 conditioned media removes the monocyte-derived fibrocyte differentiation-inhibiting activity. LGALS3BP inhibits the differentiation of monocyte-derived fibrocytes from wild-type mouse spleen cells, but not from SIGN-R1(-/-) mouse spleen cells, suggesting that CD209/SIGN-R1 is required for the LGALS3BP effect. Galectin-3 and galectin-1, binding partners of LGALS3BP, potentiate monocyte-derived fibrocyte differentiation. In breast cancer biopsies, increased levels of tumor cell-associated LGALS3BP were observed in regions of the tumor that were invading the surrounding stroma. These findings suggest LGALS3BP and galectin-3 as new targets to treat metastatic cancer and fibrosing diseases.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/pharmacology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/pharmacology
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Carrier Proteins/pharmacology
- Cell Adhesion Molecules/metabolism
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Culture Media, Conditioned
- Female
- Galectin 1/metabolism
- Galectin 3/metabolism
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Glycoproteins/pharmacology
- Humans
- Lectins, C-Type/metabolism
- Mice
- Monocytes/cytology
- Monocytes/drug effects
- Monocytes/metabolism
- RNA, Messenger/genetics
- Receptors, Cell Surface/metabolism
- Recombinant Proteins/pharmacology
Collapse
Affiliation(s)
- Michael J V White
- Department of Biology, Texas A&M University, College Station, TX 77843; and
| | - David Roife
- Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843; and
| |
Collapse
|
19
|
Shah SH, Miller P, Garcia-Contreras M, Ao Z, Machlin L, Issa E, El-Ashry D. Hierarchical paracrine interaction of breast cancer associated fibroblasts with cancer cells via hMAPK-microRNAs to drive ER-negative breast cancer phenotype. Cancer Biol Ther 2015; 16:1671-81. [PMID: 26186233 DOI: 10.1080/15384047.2015.1071742] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Multiple juxtacrine and paracrine interactions occur between cancer cells and non-cancer cells of the tumor microenvironment (TME) that direct tumor progression. Cancer Associated Fibroblasts (CAFs) are an integral component of the TME, and the majority of breast tumor stroma is comprised of CAFs. Heterotypic interactions between cancer cells and non-cancer cells of the TME occur via soluble agents, including cytokines, hormones, growth factors, and secreted microRNAs. We previously identified a microRNA signature indicative of hyperactive MAPK signaling (hMAPK-miRNA signature) that significantly associated with reduced recurrence-free and overall survival. Here we report that the hMAPK-miRNA signature associates with a high metric of stromal cell infiltrate, and we investigate the role of microRNAs, particularly hMAPK-microRNAs, secreted by CAFs on estrogen receptor (ER) expression in breast cancer cells. ER-positive MCF-7/ltE2- cells were treated with conditioned media (CM) from CAFs derived from breast cancers of different PAM50 subtypes (CAFBAS, CAFHER2, and CAFLA). CAF CM isolated specifically from ER-negative primary breast tumors led to ER repression in vitro. Nanoparticle tracking analysis and transmission electron microscopy confirmed the presence of CAF-secreted exosomes in CM and the uptake of these exosomes by the ER+ MCF-7/ltE2- cells. Differentially expressed microRNAs in CAF CM as well as in MCF-7/ltE2- cells treated with this CM were identified. Knockdown of miR-221/222 in CAFBAS resulted in knockdown of miR221/222 levels in the conditioned media and the CM from CAFBAS; miR221/222 knockdown rescued ER repression in ER-positive cell lines treated with CAFBAS-CM. Collectively, our results demonstrate that CAF-secreted microRNAs are directly involved in ER-repression, and may contribute to the MAPK-induced ER repression in breast cancer cells.
Collapse
Affiliation(s)
- Sanket H Shah
- a Cancer Biology; University of Miami ; Miami , FL USA
| | - Philip Miller
- c Sylvester Comprehensive Cancer Center; University of Miami Miller School of Medicine ; Miami , FL USA
| | - Marta Garcia-Contreras
- d Diabetes Research Institute; University of Miami Miller School of Medicine ; Miami , FL USA
| | - Zheng Ao
- a Cancer Biology; University of Miami ; Miami , FL USA
| | - Leah Machlin
- c Sylvester Comprehensive Cancer Center; University of Miami Miller School of Medicine ; Miami , FL USA
| | - Emilio Issa
- e Department of Biology ; University of Miami ; Miami , FL USA
| | - Dorraya El-Ashry
- b Department of Internal Medicine ; University of Miami Miller School of Medicine ; Miami , FL USA.,c Sylvester Comprehensive Cancer Center; University of Miami Miller School of Medicine ; Miami , FL USA
| |
Collapse
|
20
|
Arruda Macêdo JK, Fox JW, de Souza Castro M. Disintegrins from snake venoms and their applications in cancer research and therapy. Curr Protein Pept Sci 2015; 16:532-48. [PMID: 26031306 PMCID: PMC4997955 DOI: 10.2174/1389203716666150515125002] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/17/2015] [Accepted: 05/13/2015] [Indexed: 01/01/2023]
Abstract
Integrins regulate diverse functions in cancer pathology and in tumor cell development and contribute to important processes such as cell shape, survival, proliferation, transcription, angiogenesis, migration, and invasion. A number of snake venom proteins have the ability to interact with integrins. Among these are the disintegrins, a family of small, non-enzymatic, and cysteine-rich proteins found in the venom of numerous snake families. The venom proteins may have a potential role in terms of novel therapeutic leads for cancer treatment. Disintegrin can target specific integrins and as such it is conceivable that they could interfere in important processes involved in carcinogenesis, tumor growth, invasion and migration. Herein we present a survey of studies involving the use of snake venom disintegrins for cancer detection and treatment. The aim of this review is to highlight the relationship of integrins with cancer and to present examples as to how certain disintegrins can detect and affect biological processes related to cancer. This in turn will illustrate the great potential of these molecules for cancer research. Furthermore, we also outline several new approaches being created to address problems commonly associated with the clinical application of peptide-based drugs such as instability, immunogenicity, and availability.
Collapse
Affiliation(s)
| | - Jay W Fox
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, USA.
| | | |
Collapse
|
21
|
Ivers LP, Cummings B, Owolabi F, Welzel K, Klinger R, Saitoh S, O'Connor D, Fujita Y, Scholz D, Itasaki N. Dynamic and influential interaction of cancer cells with normal epithelial cells in 3D culture. Cancer Cell Int 2014; 14:108. [PMID: 25379014 PMCID: PMC4221723 DOI: 10.1186/s12935-014-0108-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/15/2014] [Indexed: 02/04/2023] Open
Abstract
Background The cancer microenvironment has a strong impact on the growth and dynamics of cancer cells. Conventional 2D culture systems, however, do not reflect in vivo conditions, impeding detailed studies of cancer cell dynamics. This work aims to establish a method to reveal the interaction of cancer and normal epithelial cells using 3D time-lapse. Methods GFP-labelled breast cancer cells, MDA-MB-231, were co-cultured with mCherry-labelled non-cancerous epithelial cells, MDCK, in a gel matrix. In the 3D culture, the epithelial cells establish a spherical morphology (epithelial sphere) thus providing cancer cells with accessibility to the basal surface of epithelia, similar to the in vivo condition. Cell movement was monitored using time-lapse analyses. Ultrastructural, immunocytochemical and protein expression analyses were also performed following the time-lapse study. Results In contrast to the 2D culture system, whereby most MDA-MB-231 cells exhibit spindle-shaped morphology as single cells, in the 3D culture the MDA-MB-231 cells were found to be single cells or else formed aggregates, both of which were motile. The single MDA-MB-231 cells exhibited both round and spindle shapes, with dynamic changes from one shape to the other, visible within a matter of hours. When co-cultured with epithelial cells, the MDA-MB-231 cells displayed a strong attraction to the epithelial spheres, and proceeded to surround and engulf the epithelial cell mass. The surrounded epithelial cells were eventually destroyed, becoming debris, and were taken into the MDA-MB-231 cells. However, when there was a relatively large population of normal epithelial cells, the MDA-MB-231 cells did not engulf the epithelial spheres effectively, despite repeated contacts. MDA-MB-231 cells co-cultured with a large number of normal epithelial cells showed reduced expression of monocarboxylate transporter-1, suggesting a change in the cell metabolism. A decreased level of gelatin-digesting ability as well as reduced production of matrix metaroproteinase-2 was also observed. Conclusions This culture method is a powerful technique to investigate cancer cell dynamics and cellular changes in response to the microenvironment. The method can be useful for various aspects such as; different combinations of cancer and non-cancer cell types, addressing the organ-specific affinity of cancer cells to host cells, and monitoring the cellular response to anti-cancer drugs. Electronic supplementary material The online version of this article (doi:10.1186/s12935-014-0108-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura P Ivers
- School of Medicine and Medical Science, University College Dublin, Dublin, 4 Ireland
| | - Brendan Cummings
- School of Medicine and Medical Science, University College Dublin, Dublin, 4 Ireland
| | - Funke Owolabi
- School of Medicine and Medical Science, University College Dublin, Dublin, 4 Ireland
| | | | - Rut Klinger
- Conway Institute, University College Dublin, Dublin, 4 Ireland ; School of Biomolecular and Biomedical Science, University College Dublin, Dublin, 4 Ireland
| | - Sayaka Saitoh
- Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815 Japan
| | - Darran O'Connor
- Conway Institute, University College Dublin, Dublin, 4 Ireland ; School of Biomolecular and Biomedical Science, University College Dublin, Dublin, 4 Ireland
| | - Yasuyuki Fujita
- Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815 Japan
| | - Dimitri Scholz
- Conway Institute, University College Dublin, Dublin, 4 Ireland
| | - Nobue Itasaki
- School of Medicine and Medical Science, University College Dublin, Dublin, 4 Ireland
| |
Collapse
|
22
|
Chatterjee SJ, McCaffrey L. Emerging role of cell polarity proteins in breast cancer progression and metastasis. BREAST CANCER-TARGETS AND THERAPY 2014; 6:15-27. [PMID: 24648766 PMCID: PMC3929326 DOI: 10.2147/bctt.s43764] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Breast cancer is a heterogeneous group of diseases that frequently exhibits loss of growth control, and disrupted tissue organization and differentiation. Several recent studies indicate that apical–basal polarity provides a tumor-suppressive function, and that disrupting polarity proteins affects many stages of breast cancer progression from initiation through metastasis. In this review we highlight some of the recent advances in our understanding of the molecular mechanisms by which loss of apical–basal polarity deregulates apoptosis, proliferation, and promotes invasion and metastasis in breast cancer.
Collapse
Affiliation(s)
- Sudipa June Chatterjee
- Rosalind and Morris Goodman Cancer Centre, Department of Oncology, McGill University, Montreal, QC, Canada
| | - Luke McCaffrey
- Rosalind and Morris Goodman Cancer Centre, Department of Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|
23
|
Hlatky L, Hahnfeldt P. Beyond the cancer cell: progression-level determinants highlight the multiscale nature of carcinogenesis risk. Cancer Res 2013; 74:659-64. [PMID: 24272486 DOI: 10.1158/0008-5472.can-13-2508] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Over the last several decades, improved awareness of the prevalence of carcinogens in the environment, along with a growing appreciation of the complexity of the carcinogenesis process, has shifted policy on cancer risk from one of strict avoidance of carcinogens to one of adherence to exposure limits deemed "safe" based on quantitative risk estimation. Meanwhile, given the mutagenic nature of most carcinogens, attention has gravitated to developing a genetic rationale for measuring and comparing risks. This focus has culminated in the now well-established multistage mutational paradigm, which holds that a stepwise sequence of mutations drives cell "initiation" and the subsequent "transformation" of an initiated cell into a cancer cell, and that, once created, a cancer cell will inevitably undergo "progression" to become overt disease. Unanticipated by this paradigm is the effect progression-phase population- and tissue-level bottleneck events may have on this process. Attesting to this is the prevalence of tumor dormancy, a state of arrested growth of an otherwise fully malignant, often microscopic cancer mass, maintained by interactions among cancer cells and between cancer and host cells. The proper inclusion of such progression-modifying influences would clearly behoove risk estimation and improve our understanding of the natural history of cancer by accounting for the less-than-certain risk of eventual cancer disease even when cancer cells are present. Such an improved understanding, in turn, stands to better inform policy-making and influence such clinical practice decisions as whether to treat the increasingly smaller tumors detectable with advancing technologies.
Collapse
Affiliation(s)
- Lynn Hlatky
- Authors' Affiliation: Center of Cancer Systems Biology, Genesys Research Institute, Tufts University School of Medicine, Boston, Massachusetts
| | | |
Collapse
|
24
|
Sun X, Gierach GL, Sandhu R, Williams T, Midkiff BR, Lissowska J, Wesolowska E, Boyd NF, Johnson NB, Figueroa JD, Sherman ME, Troester MA. Relationship of mammographic density and gene expression: analysis of normal breast tissue surrounding breast cancer. Clin Cancer Res 2013; 19:4972-4982. [PMID: 23918601 DOI: 10.1158/1078-0432.ccr-13-0029] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Previous studies of breast tissue gene expression have shown that the extratumoral microenvironment has substantial variability across individuals, some of which can be attributed to epidemiologic factors. To evaluate how mammographic density and breast tissue composition relate to extratumoral microenvironment gene expression, we used data on 121 patients with breast cancer from the population-based Polish Women's Breast Cancer Study. EXPERIMENTAL DESIGN Breast cancer cases were classified on the basis of a previously reported, biologically defined extratumoral gene expression signature with two subtypes: an Active subtype, which is associated with high expression of genes related to fibrosis and wound response, and an Inactive subtype, which has high expression of cellular adhesion genes. Mammographic density of the contralateral breast was assessed using pretreatment mammograms and a quantitative, reliable computer-assisted thresholding method. Breast tissue composition was evaluated on the basis of digital image analysis of tissue sections. RESULTS The Inactive extratumoral subtype was associated with significantly higher percentage mammographic density (PD) and dense area (DA) in univariate analysis (PD: P = 0.001; DA: P = 0.049) and in multivariable analyses adjusted for age and body mass index (PD: P = 0.004; DA: P = 0.049). Inactive/higher mammographic density tissue was characterized by a significantly higher percentage of stroma and a significantly lower percentage of adipose tissue, with no significant change in epithelial content. Analysis of published gene expression signatures suggested that Inactive/higher mammographic density tissue expressed increased estrogen response and decreased TGF-β signaling. CONCLUSIONS By linking novel molecular phenotypes with mammographic density, our results indicate that mammographic density reflects broad transcriptional changes, including changes in both epithelia- and stroma-derived signaling.
Collapse
Affiliation(s)
- Xuezheng Sun
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gretchen L Gierach
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Rupninder Sandhu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tyisha Williams
- Department of Biology, Trinity University, San Antonio, TX, USA
| | - Bentley R Midkiff
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jolanta Lissowska
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Ewa Wesolowska
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Norman F Boyd
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Toronto, ON, Canada
| | - Nicole B Johnson
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jonine D Figueroa
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mark E Sherman
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
25
|
Xu C, Wang H, He H, Zheng F, Chen Y, Zhang J, Lin X, Ma D, Zhang H. Low expression of TFPI-2 associated with poor survival outcome in patients with breast cancer. BMC Cancer 2013; 13:118. [PMID: 23497249 PMCID: PMC3607852 DOI: 10.1186/1471-2407-13-118] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 03/12/2013] [Indexed: 01/08/2023] Open
Abstract
Background The purpose of this study is to evaluate the prognostic value of TFPI-2 expression in breast cancer patients through examining the correlation between TFPI-2 expression and breast cancer clinicopathologic features. Methods Immunohistochemical staining combined with digital image analysis was used to quantify the expression of TFPI-2 protein in breast tumor tissues. For evaluation of the prognostic value of TFPI-2 expression to each clinicopathologic factor, Kaplan-Meier method and COX’s Proportional Hazard Model were employed. Results TFPI-2 expression was significantly correlated with tumor size, lymph node metastasis, histologic grade, clinical stage, and vessel invasion. More importantly, TFPI-2 expression was also associated with disease-free survival (DFS) of breast cancer patients. We found that patients with high TFPI-2 expression had longer DFS compared with those with low or negative expression of TFPI-2 (P <0.05, log-rank test). Cox’s regression analysis indicated that TFPI-2 expression, histologic grade, and vessel invasion might be significant prognostic factors for DFS, while TFPI-2 expression and histologic grade were the most significant independent predictors for tumor recurrence. Compared with the group with low/high TFPI-2 expression, the TFPI-2 negative group was more likely to have tumor relapse. The hazard ratio of DFS is 0.316 (P <0.01). Conclusions Low or negative expression of TFPI-2 is associated with breast cancer progression, recurrence and poor survival outcome after breast cancer surgery. TFPI-2 expression in breast tumors is a potential prognostic tool for breast cancer patients.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Breast Surgery, Yangpu Hospital, Tongji University, Shanghai 200090, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Artacho-Cordón A, Artacho-Cordón F, Ríos-Arrabal S, Calvente I, Núñez MI. Tumor microenvironment and breast cancer progression: a complex scenario. Cancer Biol Ther 2012; 13:14-24. [PMID: 22336584 DOI: 10.4161/cbt.13.1.18869] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It is now widely accepted that the development and progression of a tumor toward the malignant phenotype is highly dependent on interactions between tumor cells and the tumor microenvironment. Different components of the tumor microenvironment may have stimulatory or inhibitory effects on tumor progression by regulating the gene expression repertoire in tumor cells and stromal cells. This review analyzes novel research findings on breast cancer progression, discussing acquisition of the metastatic phenotype in breast disease in relation to different aspects of cross-talk among components of the tumor microenvironment. Knowledge of the interaction of all of these factors would contribute to elucidating the mechanisms that disrupt regulatory/signaling cascades and downstream effects in breast cancer.
Collapse
|
28
|
Mast cells promote the growth of Hodgkin's lymphoma cell tumor by modifying the tumor microenvironment that can be perturbed by bortezomib. Leukemia 2012; 26:2269-76. [PMID: 22430634 DOI: 10.1038/leu.2012.81] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hodgkin's lymphoma is frequently associated with mast cell infiltration that correlates directly with disease severity, but the mechanisms underlying this relationship remain unclear. Here, we report that mast cells promote the growth of Hodgkin's tumor by modifying the tumor microenvironment. A transplantation assay shows that primary murine mast cells accelerate tumor growth by established Hodgkin's cell lines, and promote marked neovascularization and fibrosis. Both mast cells and Hodgkin's cells were sensitive to bortezomib, but mast cells were more resistant to bortezomib. However, bortezomib inhibited degranulation, PGE(2)-induced rapid release of CCL2, and continuous release of vascular endothelial growth factor-A from mast cells even at the concentration that did not induce cell death. Bortezomib-treated mast cells lost the ability to induce neovasculization and fibrosis, and did not promote the growth of Hodgkin tumor in vivo. These results provide further evidence supporting causal relationships between inflammation and tumor growth, and demonstrate that bortezomib can target the tumor microenvironment.
Collapse
|
29
|
Rothberg JM, Sameni M, Moin K, Sloane BF. Live-cell imaging of tumor proteolysis: impact of cellular and non-cellular microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1824:123-32. [PMID: 21854877 PMCID: PMC3232330 DOI: 10.1016/j.bbapap.2011.07.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 07/28/2011] [Accepted: 07/29/2011] [Indexed: 01/26/2023]
Abstract
Our laboratory has had a longstanding interest in how the interactions between tumors and their microenvironment affect malignant progression. Recently, we have focused on defining the proteolytic pathways that function in the transition of breast cancer from the pre-invasive lesions of ductal carcinoma in situ (DCIS) to invasive ductal carcinomas (IDCs). We use live-cell imaging to visualize, localize and quantify proteolysis as it occurs in real-time and thereby have established roles for lysosomal cysteine proteases both pericellularly and intracellularly in tumor proteolysis. To facilitate these studies, we have developed and optimized 3D organotypic co-culture models that recapitulate the in vivo interactions of mammary epithelial cells or tumor cells with stromal and inflammatory cells. Here we will discuss the background that led to our present studies as well as the techniques and models that we employ. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cells, Cultured
- Cellular Microenvironment/physiology
- Diagnostic Imaging/methods
- Female
- Humans
- Microscopy, Video
- Models, Biological
- Neoplasms/diagnosis
- Neoplasms/metabolism
- Neoplasms/pathology
- Proteolysis
- Single-Cell Analysis/methods
- Tumor Microenvironment/physiology
Collapse
Affiliation(s)
- Jennifer M Rothberg
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
30
|
Páez D, Labonte MJ, Bohanes P, Zhang W, Benhanim L, Ning Y, Wakatsuki T, Loupakis F, Lenz HJ. Cancer dormancy: a model of early dissemination and late cancer recurrence. Clin Cancer Res 2011; 18:645-53. [PMID: 22156560 DOI: 10.1158/1078-0432.ccr-11-2186] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer dormancy is a stage in tumor progression in which residual disease remains occult and asymptomatic for a prolonged period of time. Dormant tumor cells can be present as one of the earliest stages in tumor development, as well as a stage in micrometastases, and/or minimal residual disease left after an apparently successful treatment of the primary tumor. The general mechanisms that regulate the transition of disseminated tumor cells that have lain dormant into a proliferative state remain largely unknown. However, regulation of the growth from dormant tumor cells may be explained in part through the interaction of the tumor cell with its microenvironment, limitations in the blood supply, or an active immune system. An understanding of the regulatory machinery of these processes is essential for identifying early cancer biomarkers and could provide a rationale for the development of novel agents to target dormant tumor cells. This review focuses on the different signaling models responsible for early cancer dissemination and tumor recurrence that are involved in dormancy pathways.
Collapse
Affiliation(s)
- David Páez
- Division of Medical Oncology and USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chandrasekaran S, Giang UBT, King MR, DeLouise LA. Microenvironment induced spheroid to sheeting transition of immortalized human keratinocytes (HaCaT) cultured in microbubbles formed in polydimethylsiloxane. Biomaterials 2011; 32:7159-68. [PMID: 21724250 DOI: 10.1016/j.biomaterials.2011.06.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 06/07/2011] [Indexed: 11/24/2022]
Abstract
The in vivo cellular microenvironment is regulated by a complex interplay of soluble factors and signaling molecules secreted by cells and it plays a critical role in the growth and development of normal and diseased tissues. In vitro systems that can recapitulate the microenvironment at the cellular level are needed to investigate the influence of autocrine signaling and extracellular matrix effects on tissue homeostasis, regeneration, disease development and progression. In this study, we report the use of microbubble technology as a means to culture cells in a controlled microenvironment in which cells can influence their function through autocrine signaling. Microbubbles (MB) are small spherical cavities about 100-300 μm in diameter formed in hydrophobic polydimethylsiloxane (PDMS) with ∼60-100 μm circular openings and aspect ratio ∼3.0. We demonstrate that the unique architecture of the microbubble compartment is advantaged for cell culture using HaCaT cells, an immortalized keratinocyte cell line. We observe that HaCaT cells, seeded in microbubbles (15-20 cells/MB) and cultured under standard conditions, adopt a compact 3D spheroidal morphology. Within 2-3 days, the cells transition to a sheeting morphology. Through experimentation and simulation we show that this transition in morphology is due to the unique architecture of the microbubble compartment which enables cells to condition their local microenvironment. The small media volume per cell and the development of shallow concentration gradients allow factors secreted by the cells to rise to bioactive levels. The kinetics of the morphology transition depends on the number of cells seeded per microbubble; higher cell seeding induces a more rapid transition. HaCaT cells seeded onto PDMS cured in 96-well plates also form compact spheroids but they do not undergo a transition to a sheeting morphology even after several weeks of culture. The importance of soluble factor accumulation in driving this morphology transition in microbubbles is supported by the observation that spheroids do not form when cells - seeded into microbubbles or onto PDMS cured in 96-well plates - are cultured in media conditioned by HaCaT cells grown in standard tissue culture plate. We observed that the addition of TGF-β1 to the growth media induced cells to proliferate in a sheeting morphology from the onset both on PDMS cured in 96-well plates and in microbubbles. TGF-β1 is a morphogen known to regulate epithelial-to-mesenchymal transition (EMT). Studies of the role of Ca(2+) concentration and changes in E-cadherin expression additionally support an EMT-like HaCaT morphology transition. These findings taken together validate the microbubble compartment as a unique cell culture platform that can potentially transform investigative studies in cell biology and in particular the tumor microenvironment. Targeting the tumor microenvironment is an emerging area of anti-cancer therapy.
Collapse
|
32
|
Tartakover-Matalon S, Mizrahi A, Epstein G, Shneifi A, Drucker L, Pomeranz M, Fishman A, Radnay J, Lishner M. Breast cancer characteristics are modified by first trimester human placenta: in vitro co-culture study. Hum Reprod 2010; 25:2441-54. [PMID: 20719812 DOI: 10.1093/humrep/deq227] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pregnant women with breast cancer present with a more advanced disease compared with non-pregnant women. Nevertheless, breast cancer metastasis to the placenta is rare. Trophoblast/tumor implantations share the same biochemical mediators, while only the first is stringently controlled. We hypothesized that the same mechanisms that affect/restrain placental implantation may inhibit metastatic growth in the placenta. We aimed to analyze the effects of human placenta on breast cancer cells. METHODS First trimester human placental explants were co-cultured with MCF-7/T47D-eGFP tagged cells. Following culture, placenta/cancer cells/both were fixed, paraffin embedded and sliced for immunohistochemical analysis or sorted by their eGFP expression for future analysis. The tested parameters were: proliferation (immunohistochemistry)/cell cycle (FACS), apoptosis (immunohistochemistry/FACS), cell count/adhesion/distribution around the placenta (cell sorter, visual observation and counting), matrix metalloproteinase activity (zymogram) and estrogen receptor (ER) expression (western blotting, immunohistochemistry). RESULTS Reduced breast cancer cell numbers (45%↓, 48%↓ for MCF-7/T47D, respectively, P < 0.05) were observed near the placenta. The placenta elevated MCF-7 sub-G1 phase and modestly elevated apoptosis (3-17%↑ for T47D/MCF-7, respectively, P < 0.05). Our findings demonstrate breast cancer cell migration from the placenta as: (i) T47D/MCF-7 cells changed their morphology to that of motile cells; (ii) elevated MMPs activity was found in the co-culture; (iii) placental soluble factors detached breast cancer cells; and (4) the placenta reduced MCF-7/T47D cells' ER expression (a characteristic of motile cells). CONCLUSIONS MCF-7/T47D cells are eliminated from the placental surroundings. Analyzing the causes of these phenomena may suggest biological pathways for this event and raise new therapeutic targets.
Collapse
Affiliation(s)
- S Tartakover-Matalon
- Oncogenetic Laboratory, Meir Medical Center, 45 Tschernehovski St, Kfar Saba 44281, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Barkan D, El Touny LH, Michalowski AM, Smith JA, Chu I, Davis AS, Webster JD, Hoover S, Simpson RM, Gauldie J, Green JE. Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res 2010; 70:5706-16. [PMID: 20570886 DOI: 10.1158/0008-5472.can-09-2356] [Citation(s) in RCA: 289] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer that recurs as metastatic disease many years after primary tumor resection and adjuvant therapy seems to arise from tumor cells that disseminated early in the course of disease but did not develop into clinically apparent lesions. These long-term surviving, disseminated tumor cells maintain a state of dormancy, but may be triggered to proliferate through largely unknown factors. We now show that the induction of fibrosis, associated with deposition of type I collagen (Col-I) in the in vivo metastatic microenvironment, induces dormant D2.0R cells to form proliferative metastatic lesions through beta1-integrin signaling. In vitro studies using a three-dimensional culture system modeling dormancy showed that Col-I induces quiescent D2.0R cells to proliferate through beta1-integrin activation of SRC and focal adhesion kinase, leading to extracellular signal-regulated kinase (ERK)-dependent myosin light chain phosphorylation by myosin light chain kinase and actin stress fiber formation. Blocking beta1-integrin, Src, ERK, or myosin light chain kinase by short hairpin RNA or pharmacologic approaches inhibited Col-I-induced activation of this signaling cascade, cytoskeletal reorganization, and proliferation. These findings show that fibrosis with Col-I enrichment at the metastatic site may be a critical determinant of cytoskeletal reorganization in dormant tumor cells, leading to their transition from dormancy to metastatic growth. Thus, inhibiting Col-I production, its interaction with beta1-integrin, and downstream signaling of beta1-integrin may be important strategies for preventing or treating recurrent metastatic disease.
Collapse
Affiliation(s)
- Dalit Barkan
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
O'Brien J, Lyons T, Monks J, Lucia MS, Wilson RS, Hines L, Man YG, Borges V, Schedin P. Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1241-55. [PMID: 20110414 DOI: 10.2353/ajpath.2010.090735] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Recent pregnancy correlates with decreased survival for breast cancer patients compared with non-pregnancy-associated breast cancer. We hypothesize that postpartum mammary involution induces metastasis through wound-healing programs known to promote cancer. It is unknown whether alternatively activated M2 macrophages, immune cells important in wound-healing and experimental tumorigenesis that also predict poor prognosis for breast cancer patients, are recruited to the normal involuting gland. Macrophage markers CD68, CSF-1R, and F4/80 were examined across the pregnancy and involution cycle in rodent and human mammary tissues. Quantitative immunohistochemistry revealed up to an eightfold increase in macrophage number during involution, which returned to nulliparous levels with full regression. The involution macrophages exhibit an M2 phenotype as determined by high arginase-1 and low inducible nitric oxide synthase staining in rodent tissue, and by mannose receptor expression in human breast tissue. M2 cytokines IL-4 and IL-13 also peaked during involution. Extracellular matrix (ECM) isolated from involuting rat mammary glands was chemotactic for macrophages compared with nulliparous mammary ECM. Fibrillar collagen levels and proteolysis increased dramatically during involution, and denatured collagen I acted as a strong chemoattractant for macrophages in cell culture, suggesting proteolyzed fibrillar collagen as a candidate ECM mediator of macrophage recruitment. M2 macrophages, IL-4, IL-13, fibrillar collagen accumulation, and proteolysis of collagen are all components of tumor promotional microenvironments, and thus may mediate promotion of breast cancers arising in the postpartum setting.
Collapse
Affiliation(s)
- Jenean O'Brien
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, Mail Stop 8117, RC-1 S, 8401K, 12801 E. 17 Avenue, UCD, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
The clinical implications of MMP-11 and CK-20 expression in human breast cancer. Clin Chim Acta 2009; 411:234-41. [PMID: 19914229 DOI: 10.1016/j.cca.2009.11.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 11/08/2009] [Accepted: 11/09/2009] [Indexed: 12/21/2022]
Abstract
BACKGROUND Tumor invasiveness and metastasis in cancer progression is manifested by epigenetic abnormality. However, it remains unknown whether transcription regulation of matrix metalloproteinase-11(MMP-11) and cytoskeleton-20 (CK-20) genes for the homoeostasis of epithelial/connective interface that can enhance cell dissemination and invasion may act as alternative mutators to tumor clinicopathology. METHODS Paired cancerous and tumor-adjacent normal tissues from 72 breast cancer patients were assayed for the expression of MMP-11 and CK-20 by using real-time RT-PCR. The expression profiles were evaluated for the association with clinicopathological factors. RESULTS Breast tumor tissues displayed higher expression levels of MMP-11 and CK-20 than those of the adjacent non-cancerous tissues. Overexpression of either MMP-11 or CK-20 correlated with patients having poorly differentiated tumors (P(MMP-11)=0.01 and P(CK-20)=0.05) and lymph node metastasis (LNM) (P(MMP-11)=0.004 and P(CK-20)=0.001). A synergistic effect between MMP-11 and CK-20 on risk elevation was significant in patients with advanced tumor stage (OR=2.03, 95%CI=1.10-3.77) and LNM (OR=2.83, 95%CI=1.20-4.71). Additionally, patients lacking progesterone receptor exhibited high expression of MMP-11 and CK-20. CONCLUSION We demonstrate that MMP-11 and CK-20 are probable prognostic markers whose expression reflects the stages of tumor differentiation and LNM of breast cancer.
Collapse
|
36
|
Mukherjee S, Richardson AM, Rodriguez-Canales J, Ylaya K, Erickson HS, Player A, Kawasaki ES, Pinto PA, Choyke PL, Merino MJ, Albert PS, Chuaqui RF, Emmert-Buck MR. Identification of EpCAM as a molecular target of prostate cancer stroma. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:2277-87. [PMID: 19850885 DOI: 10.2353/ajpath.2009.090013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
To delineate the molecular changes that occur in the tumor microenvironment, we previously performed global transcript analysis of human prostate cancer specimens using tissue microdissection and expression microarrays. Epithelial and stromal compartments were individually studied in both tumor and normal fields. Tumor-associated stroma showed a distinctly different expression pattern compared with normal stroma, having 44 differentially expressed transcripts, the majority of which were up-regulated. In the present study, one of the up-regulated transcripts, epithelial cell adhesion activating molecule, was further evaluated at the protein level in 20 prostate cancer cases using immunohistochemistry and a histomathematical analysis strategy. The epithelial cell adhesion activating molecule showed a 76-fold expression increase in the tumor-associated stroma, as compared with matched normal stroma. Moreover, Gleason 4 or 5 tumor stroma was increased 170-fold relative to matched normal stroma, whereas the Gleason 3 tumor area showed only a 36-fold increase, indicating a positive correlation with Gleason tumor grade. Since the stromal compartment may be particularly accessible to vascular-delivered agents, epithelial cell adhesion activating molecule could become a valuable molecular target for imaging or treatment of prostate cancer.
Collapse
Affiliation(s)
- Sumana Mukherjee
- Pathogenetics Unit, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4605, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hansen KC, Kiemele L, Maller O, O'Brien J, Shankar A, Fornetti J, Schedin P. An in-solution ultrasonication-assisted digestion method for improved extracellular matrix proteome coverage. Mol Cell Proteomics 2009; 8:1648-57. [PMID: 19351662 DOI: 10.1074/mcp.m900039-mcp200] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Epithelial cell behavior is coordinated by the composition of the surrounding extracellular matrix (ECM); thus ECM protein identification is critical for understanding normal biology and disease states. Proteomic analyses of ECM proteins have been hindered by the insoluble and digestion-resistant nature of ECM. Here we explore the utility of combining rapid ultrasonication- and surfactant-assisted digestion for the detailed proteomics analysis of ECM samples. When compared with traditional overnight digestion, this optimized method dramatically improved the sequence coverage for collagen I, revealed the presence of hundreds of previously unidentified proteins in Matrigel, and identified a protein profile for ECM isolated from rat mammary glands that was substantially different from that found in Matrigel. In a three-dimensional culture assay to investigate epithelial cell-ECM interactions, mammary epithelial cells were found to undergo extensive branching morphogenesis when plated with mammary gland-derived matrix in comparison with Matrigel. Cumulatively these data highlight the tissue-specific nature of ECM composition and function and underscore the need for optimized techniques, such as those described here, for the proteomics characterization of ECM samples.
Collapse
Affiliation(s)
- Kirk C Hansen
- University of Colorado Cancer Center Proteomics and Mass Spectrometry Facility, Department of Pediatrics, University of Colorado Denver, Aurora, Colorado 80045, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Schedin P, Borges V. Breaking down barriers: the importance of the stromal microenvironment in acquiring invasiveness in young women's breast cancer. Breast Cancer Res 2009; 11:102. [PMID: 19344495 PMCID: PMC2688940 DOI: 10.1186/bcr2235] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gene expression profiling was performed on laser captured breast stroma and epithelium obtained from 14 breast cancer patients. As with breast epithelium, of the stromal gene expression changes observed between normal tissue and invasive ductal carcinoma, greater than 90% occurred early, at the normal to ductal carcinoma in situ transition. Only 10% of stromal and 0% of epithelial genes were differentially regulated between non-invasive ductal carcinoma in situ and invasive disease. These data suggest that the majority of gene expression changes required for transformation occur early, prior to histological evidence of an invasive phenotype, the stroma cooperates closely with epithelium in this transformation, and for acquisition of the invasive phenotype, the stroma is dominant over the epithelium.
Collapse
Affiliation(s)
- Pepper Schedin
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, Aurora, CO 80045, USA.
| | | |
Collapse
|
39
|
Arnold JT. DHEA metabolism in prostate: For better or worse? Mol Cell Endocrinol 2009; 301:83-8. [PMID: 19013497 PMCID: PMC2667103 DOI: 10.1016/j.mce.2008.10.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 10/14/2008] [Accepted: 10/15/2008] [Indexed: 11/16/2022]
Abstract
Dehydroepiandrosterone (DHEA) is commonly used in the USA as a nutritional supplement for antiaging, metabolic support or other uses. Investigations into understanding the effects of DHEA on human prostate cancer progression have posed more questions than answers and highlight the importance of communications between stromal and epithelial tuoitiuot elements within the prostate that contribute to the regulation of DHEA metabolism. Intracrine metabolism of DHEA to androgens (A) and/or estrogens (E) may occur in one cell compartment (stromal) which may release paracrine hormones or growth/inhibitory factors to the epithelial cells. Alternatively no metabolism of DHEA may occur, resulting in no harmful consequences of high levels of DHEA in prostate tissues. We herein review the tissue components involved and interactions with the prohormone, DHEA and/or resulting metabolites, including dihydrotestosterone (DHT) or 17beta-estradiol (E(2)) in an in vitro model of endocrine-immune-paracrine interactions within the prostate. This work raises questions and hypotheses concerning the role of DHEA in prostate in normal tissues, vs. preneoplastic tissues.
Collapse
Affiliation(s)
- Julia T Arnold
- LCI-Endocrine Section, National Center for Complementary and Alternative Medicine (NCCAM), National Institutes of Health (NIH), Building 10/2B47 MSC 1547, 9000 Rockville Pike, Bethesda, MD 20892-1547, USA.
| |
Collapse
|
40
|
Gray NE, Liu X, Choi R, Blackman MR, Arnold JT. Endocrine-immune-paracrine interactions in prostate cells as targeted by phytomedicines. Cancer Prev Res (Phila) 2009; 2:134-42. [PMID: 19141600 DOI: 10.1158/1940-6207.capr-08-0062] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dehydroepiandrosterone (DHEA) is used as a dietary supplement and can be metabolized to androgens and/or estrogens in the prostate. We investigated the hypothesis that DHEA metabolism may be increased in a reactive prostate stroma environment in the presence of proinflammatory cytokines such as transforming growth factor beta1 (TGFbeta1), and further, whether red clover extract, which contains a variety of compounds including isoflavones, can reverse this effect. LAPC-4 prostate cancer cells were grown in coculture with prostate stromal cells (6S) and treated with DHEA +/- TGFbeta1 or interleukin-6. Prostate-specific antigen (PSA) expression and testosterone secretion in LAPC-4/6S cocultures were compared with those in monocultured epithelial and stromal cells by real-time PCR and/or ELISA. Combined administration of TGFbeta1 + DHEA to cocultures increased PSA protein secretion two to four times, and PSA gene expression up to 50-fold. DHEA + TGFbeta1 also increased coculture production of testosterone over DHEA treatment alone. Red clover isoflavone treatment led to a dose-dependent decrease in PSA protein and gene expression and testosterone metabolism induced by TGFbeta1 + DHEA in prostate LAPC-4/6S cocultures. In this coculture model of endocrine-immune-paracrine interactions in the prostate, TGFbeta1 greatly increased stromal-mediated DHEA effects on testosterone production and epithelial cell PSA production, whereas red clover isoflavones reversed these effects.
Collapse
Affiliation(s)
- Nora E Gray
- Endocrine Section, Laboratory of Clinical Investigation, Division of Intramural Research, National Center for Complementary and Alternative Medicine, NIH, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|
41
|
Sameni M, Cavallo-Medved D, Dosescu J, Jedeszko C, Moin K, Mullins SR, Olive MB, Rudy D, Sloane BF. Imaging and quantifying the dynamics of tumor-associated proteolysis. Clin Exp Metastasis 2008; 26:299-309. [PMID: 19082919 PMCID: PMC2991638 DOI: 10.1007/s10585-008-9218-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Accepted: 09/26/2008] [Indexed: 12/16/2022]
Abstract
The roles of proteases in cancer are dynamic. Furthermore, the roles or functions of any one protease may differ from one stage of cancer to another. Proteases from tumor-associated cells (e.g., fibroblasts, inflammatory cells, endothelial cells) as well as from tumor cells make important contributions to 'tumor proteolysis'. Many tumors exhibit increases in expression of proteases at the level of transcripts and protein; however, whether those proteases play causal roles in malignant progression is known for only a handful of proteases. What the critical substrate or substrates that are cleaved in vivo by any given protease is also known for only a few proteases. Therefore, the recent development of techniques and reagents for live cell imaging of protease activity, in conjunction with informed knowledge of critical natural substrates, should help to define protease functions. Here we describe live cell assays for imaging proteolysis, protocols for quantifying proteolysis and the use of such assays to follow the dynamics of proteolysis by tumor cells alone and tumor cells interacting with other cells found in the tumor microenvironment. In addition, we describe an in vitro model that recapitulates the architecture of the mammary gland, a model designed to determine the effects of dynamic interactions with the surrounding microenvironment on 'tumor proteolysis' and the respective contributions of various cell types to 'tumor proteolysis'. The assays and models described here could serve as screening platforms for the identification of proteolytic pathways that are potential therapeutic targets and for further development of technologies and imaging probes for in vivo use.
Collapse
Affiliation(s)
- Mansoureh Sameni
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Dora Cavallo-Medved
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA, Barbara Ann Karmanos Cancer Institute, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Julie Dosescu
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Christopher Jedeszko
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Kamiar Moin
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA, Barbara Ann Karmanos Cancer Institute, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Stefanie R. Mullins
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA, Barbara Ann Karmanos Cancer Institute, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Mary B. Olive
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Deborah Rudy
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Bonnie F. Sloane
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA, , Barbara Ann Karmanos Cancer Institute, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| |
Collapse
|
42
|
Bennett CN, Green JE. Unlocking the power of cross-species genomic analyses: identification of evolutionarily conserved breast cancer networks and validation of preclinical models. Breast Cancer Res 2008; 10:213. [PMID: 18828875 PMCID: PMC2614501 DOI: 10.1186/bcr2125] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The application of high-throughput genomic technologies has revealed that individual breast tumors display a variety of molecular features that require more personalized approaches to treatment. Several recent studies have demonstrated that a cross-species analytic approach provides a powerful means to filter through genetic complexity by identifying evolutionarily conserved genetic networks that are fundamental to the oncogenic process. Mouse-human tumor comparisons will provide insights into cellular origins of tumor subtypes, define interactive oncogenetic networks, identify potential novel therapeutic targets, and further validate as well as guide the selection of genetically engineered mouse models for preclinical testing.
Collapse
Affiliation(s)
- Christina N Bennett
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | | |
Collapse
|
43
|
Demicheli R, Retsky MW, Hrushesky WJM, Baum M, Gukas ID. The effects of surgery on tumor growth: a century of investigations. Ann Oncol 2008; 19:1821-8. [PMID: 18550576 DOI: 10.1093/annonc/mdn386] [Citation(s) in RCA: 271] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
A few clinical investigations suggest that while primary breast cancer surgical removal favorably modifies the natural history for some patients, it may also hasten the metastatic development for others. The concepts underlying this disease paradigm, i.e. tumor homeostasis, tumor dormancy and surgery-driven enhancement of metastasis development, have a long history that is reviewed. The review reveals the context in which these concepts were conceived and structured to explain experimental data and shows that they are not so new and far fetched. The idea that surgical cancer resection has both beneficial and adverse effects upon cancer spread and growth that result from the modulation of tumor dormancy by the resection should be considered a potentially fruitful working hypothesis.
Collapse
Affiliation(s)
- R Demicheli
- Department of Medical Oncology, Istituto Nazionale Tumori, Milano, Italy.
| | | | | | | | | |
Collapse
|
44
|
Adipocyte culture medium stimulates production of macrophage inhibitory cytokine 1 in MDA-MB-231 cells. Cancer Lett 2008; 261:253-62. [DOI: 10.1016/j.canlet.2007.11.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Revised: 11/07/2007] [Accepted: 11/19/2007] [Indexed: 11/18/2022]
|
45
|
Raynaud CM, Sabatier L, Philipot O, Olaussen KA, Soria JC. Telomere length, telomeric proteins and genomic instability during the multistep carcinogenic process. Crit Rev Oncol Hematol 2008; 66:99-117. [PMID: 18243729 DOI: 10.1016/j.critrevonc.2007.11.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 11/08/2007] [Accepted: 11/30/2007] [Indexed: 12/29/2022] Open
Abstract
Telomeres form specialized structures at the ends of eukaryotic chromosomes, preventing them from being wrongly recognized as DNA damage. The human telomere DNA sequence is a tandem repetition of the sequence TTAGGG. In normal cells, the DNA replication machinery is unable to completely duplicate the telomeric DNA; thus, telomeres are shortened after every cell division. Having reached a critical length, telomeres may be recognized as double strand break DNA lesions, and cells eventually enter senescence. Carcinogenesis is a multistep process involving multiple mutations and chromosomal aberrations. One of the most prevalent aberrations in pre-cancerous lesions is telomere shortening and telomerase activation. We discuss the role and homeostasis of telomeres in normal cells and their implication in the early steps of carcinogenesis. We also discuss various techniques used, and their limitations, in the study of telomeres and genome instability and their role in carcinogenesis and related genomic modifications.
Collapse
|
46
|
Abstract
Characterization of gene expression profiles in tumor cells and the tumor microenvironment is an important step in understanding neoplastic progression. To date, there are limited data available on expression changes that occur in the tumor-associated stroma as either a cause or consequence of cancer. In the present study, we employed a 54,000 target oligonucleotide microarray to compare expression profiles in the 4 major components of the microenvironment: tumor epithelium, tumor-associated stroma, normal epithelium, and normal stroma. Cells from 5 human, whole-mount prostatectomy specimens were microdissected and the extracted and amplified mRNA was hybridized to an Affymetrix Human Genome U133 Plus 2.0 GeneChip. Using the intersection of 2 analysis methods, we identified sets of differentially expressed genes among the 4 components. Forty-four genes were found to be consistently differentially expressed in the tumor-associated stroma; 35 were found in the tumor epithelium. Interestingly, the tumor-associated stroma showed a predominant up-regulation of transcripts compared with normal stroma, in sharp contrast to the overall down-regulation seen in the tumor epithelium relative to normal epithelium. These data provide insight into the molecular changes occurring in tumor-associated stromal cells and suggest new potential targets for future diagnostic, imaging, or therapeutic intervention.
Collapse
|
47
|
Parker BS, Ciocca DR, Bidwell BN, Gago FE, Fanelli MA, George J, Slavin JL, Möller A, Steel R, Pouliot N, Eckhardt BL, Henderson MA, Anderson RL. Primary tumour expression of the cysteine cathepsin inhibitor Stefin A inhibits distant metastasis in breast cancer. J Pathol 2007; 214:337-46. [DOI: 10.1002/path.2265] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
48
|
Flavell SJ, Hou TZ, Lax S, Filer AD, Salmon M, Buckley CD. Fibroblasts as novel therapeutic targets in chronic inflammation. Br J Pharmacol 2007; 153 Suppl 1:S241-6. [PMID: 17965753 DOI: 10.1038/sj.bjp.0707487] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A characteristic feature of many chronic inflammatory diseases is their persistence and predilection for certain sites. The molecular basis for such tissue tropism and failure of the inflammatory response to resolve has until relative recently remained obscure. Recent studies have strongly implicated fibroblasts as cells which contribute to disease persistence and which help define anatomical location. Therefore fibroblasts make an attractive therapeutic target as they help orchestrate the inflammatory infiltrate. Current anti-inflammatory therapies target immune cells in an attempt to inhibit the production of pro-inflammatory mediators. However an equally important target is the active induction of pro-resolution programmes responsible for the resolution of inflammation. Fibroblasts are likely to be an important source of these anti-inflammatory mediators. Therapeutic manipulation of fibroblasts and their biologically active products is an emerging concept in treating cancer and is likely to provide a novel method to achieve improved control of chronic inflammatory disease.
Collapse
Affiliation(s)
- S J Flavell
- Rheumatology Research Group, Division of Immunity and Infection, MRC Centre for Immune Regulation, University of Birmingham, Birmingham, UK
| | | | | | | | | | | |
Collapse
|
49
|
Vecchi M, Confalonieri S, Nuciforo P, Viganò MA, Capra M, Bianchi M, Nicosia D, Bianchi F, Galimberti V, Viale G, Palermo G, Riccardi A, Campanini R, Daidone MG, Pierotti MA, Pece S, Di Fiore PP. Breast cancer metastases are molecularly distinct from their primary tumors. Oncogene 2007; 27:2148-58. [PMID: 17952122 DOI: 10.1038/sj.onc.1210858] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metastases have been widely thought to arise from rare, selected, mutation-bearing cells in the primary tumor. Recently, however, it has been proposed that breast tumors are imprinted ab initio with metastatic ability. Thus, there is a debate over whether 'phenotypic' disease progression is really associated with 'molecular' progression. We profiled 26 matched primary breast tumors and lymph node metastases and identified 270 probesets that could discriminate between the two categories. We then used an independent cohort of breast tumors (81 samples) and unmatched distant metastases (32 samples) to validate and refine this list down to a 126-probeset list. A representative subset of these genes was subjected to analysis by in situ hybridization, on a third independent cohort (57 primary breast tumors and matched lymph node metastases). This not only confirmed the expression profile data, but also allowed us to establish the cellular origin of the signals. One-third of the analysed representative genes (4 of 11) were expressed by the epithelial component. The four epithelial genes alone were able to discriminate primary breast tumors from their metastases. Finally, engineered alterations in the expression of two of the epithelial genes (SERPINB5 and LTF) modified cell motility in vitro, in accordance with a possible causal role in metastasis. Our results show that breast cancer metastases are molecularly distinct from their primary tumors.
Collapse
Affiliation(s)
- M Vecchi
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chiang GG, Abraham RT. Targeting the mTOR signaling network in cancer. Trends Mol Med 2007; 13:433-42. [PMID: 17905659 DOI: 10.1016/j.molmed.2007.08.001] [Citation(s) in RCA: 246] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 08/01/2007] [Accepted: 08/13/2007] [Indexed: 01/16/2023]
Abstract
The mammalian target of rapamycin (mTOR) is an unconventional protein kinase that is centrally involved in the control of cancer cell metabolism, growth and proliferation. The mTOR pathway has attracted broad scientific and clinical interest, particularly in light of the ongoing clinical cancer trials with mTOR inhibitors. The mixed clinical results to date reflect the complexity of both cancer as a disease target, and the mTOR signaling network, which contains two functionally distinct mTOR complexes, parallel regulatory pathways, and feedback loops that contribute to the variable cellular responses to the current inhibitors. In this review, we discuss the regulatory pathways that govern mTOR activity, and highlight clinical results obtained with the first generation of mTOR inhibitors to reach the oncology clinics.
Collapse
Affiliation(s)
- Gary G Chiang
- Program in Signal Transduction, Burnham Institute for Medical Research, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|