1
|
Das A, Giri S, Dey P. Cell-cell junctional proteins in cancer. Adv Clin Chem 2024; 125:93-142. [PMID: 39988409 DOI: 10.1016/bs.acc.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
A hallmark change during carcinogenesis is disruption or dysregulation of cell-cell junctions. It enables a transformed cell to adopt mesenchymal phenotype and acquire higher potential to migrate and invade. This ultimately leads to cancer metastasis. During this process, junctional proteins undergo remarkable changes in terms of their expressional pattern, localization, and activity. De-localized junctional proteins may adopt atypical roles which might act to either suppress tumorigenesis or facilitate cancer development, depending on several factors. In this chapter, the authors attempt to know the expression pattern of junctional proteins in different types of cancer, understand its significance, and gather knowledge about the mechanisms by which they regulate tumorigenesis and cancer development.
Collapse
Affiliation(s)
- Aparajita Das
- Molecular and Cell Biology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Sarbani Giri
- Molecular and Cell Biology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India.
| | - Pubali Dey
- Molecular and Cell Biology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| |
Collapse
|
2
|
Watanabe M, Nakai H, Ohara T, Kawasaki K, Murosaki S, Hirose Y. Beneficial effect of heat-killed Lactiplantibacillus plantarum L-137 on intestinal barrier function of rat small intestinal epithelial cells. Sci Rep 2024; 14:12319. [PMID: 38811623 PMCID: PMC11136994 DOI: 10.1038/s41598-024-62657-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
Heat-killed Lactiplantibacillus plantarum L-137 (HK L-137) has been suggested to enhance the intestinal barrier in obese mice, leading to improvement of metabolic abnormalities and adipose tissue inflammation, and in healthy humans with overweight, leading to improvement of systemic inflammation. However, its detailed mechanism of action has not been clarified. Therefore, this study investigated the effects of HK L-137 on the permeability of rat small intestinal epithelial IEC-6 cells, tight junction-related gene and protein expression and localization, and intracellular signaling pathways involved in barrier function. Treatment of IEC-6 cells with HK L-137 for 26 h significantly reduced the permeability to fluorescein isothiocyanate-dextran (FD-4). HK L-137 also increased gene and protein expression of zonula occludens-1 (ZO-1), an important tight junction protein, without affecting the localization. Furthermore, inhibition of the extracellular signal-regulated kinase (ERK)1/2 pathway in IEC-6 cells canceled the HK L-137-related reduction in permeability to FD-4. Phosphorylation of ERK in IEC-6 cells was induced 15 min after the addition of HK L-137. These results suggest that HK L-137 reduces intestinal permeability partly through activating the ERK pathway and increasing expression of the ZO-1 gene and protein. Enhancement of intestinal barrier function with HK L-137 might be effective in preventing and treating leaky gut, for which no specific therapeutic tool has been established.
Collapse
Affiliation(s)
- Mototsugu Watanabe
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan.
| | - Hiroko Nakai
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| | - Tatsuya Ohara
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| | - Kengo Kawasaki
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| | - Shinji Murosaki
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| | - Yoshitaka Hirose
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| |
Collapse
|
3
|
Hozhabri H, Ghasemi Dehkohneh RS, Razavi SM, Razavi SM, Salarian F, Rasouli A, Azami J, Ghasemi Shiran M, Kardan Z, Farrokhzad N, Mikaeili Namini A, Salari A. Comparative analysis of protein-protein interaction networks in metastatic breast cancer. PLoS One 2022; 17:e0260584. [PMID: 35045088 PMCID: PMC8769308 DOI: 10.1371/journal.pone.0260584] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 11/12/2021] [Indexed: 12/27/2022] Open
Abstract
Metastatic lesions leading causes of the majority of deaths in patients with the breast cancer. The present study aimed to provide a comprehensive analysis of the differentially expressed genes (DEGs) in the brain (MDA-MB-231 BrM2) and lung (MDA-MB-231 LM2) metastatic cell lines obtained from breast cancer patients compared with those who have primary breast cancer. We identified 981 and 662 DEGs for brain and lung metastasis, respectively. Protein-protein interaction (PPI) analysis revealed seven shared (PLCB1, FPR1, FPR2, CX3CL1, GABBR2, GPR37, and CXCR4) hub genes between brain and lung metastasis in breast cancer. Moreover, GNG2 and CXCL8, C3, and PTPN6 in the brain and SAA1 and CCR5 in lung metastasis were found as unique hub genes. Besides, five co-regulation of clusters via seven important co-expression genes (COL1A2, LUM, SPARC, THBS2, IL1B, CXCL8, THY1) were identified in the brain PPI network. Clusters screening followed by biological process (BP) function and pathway enrichment analysis for both metastatic cell lines showed that complement receptor signalling, acetylcholine receptor signalling, and gastric acid secretion pathways were common between these metastases, whereas other pathways were site-specific. According to our findings, there are a set of genes and functional pathways that mark and mediate breast cancer metastasis to the brain and lungs, which may enable us understand the molecular basis of breast cancer development in a deeper levele to the brain and lungs, which may help us gain a more complete understanding of the molecular underpinnings of breast cancer development.
Collapse
Affiliation(s)
- Hossein Hozhabri
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
| | - Roxana Sadat Ghasemi Dehkohneh
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Seyed Morteza Razavi
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - S. Mostafa Razavi
- Department of Chemical, Biomolecular and Corrosion Engineering, The University of Akron, Akron, Ohio, United States of America
| | - Fatemeh Salarian
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Azade Rasouli
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalil Azami
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Melika Ghasemi Shiran
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Biology, Faculty of Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Kardan
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Cellular Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Negar Farrokhzad
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Arsham Mikaeili Namini
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Ali Salari
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
4
|
Neyrinck-Leglantier D, Lesage J, Blacher S, Bonnomet A, Hunziker W, Noël A, Dormoy V, Nawrocki-Raby B, Gilles C, Polette M. ZO-1 Intracellular Localization Organizes Immune Response in Non-Small Cell Lung Cancer. Front Cell Dev Biol 2021; 9:749364. [PMID: 34938731 PMCID: PMC8685499 DOI: 10.3389/fcell.2021.749364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022] Open
Abstract
Delocalization of zonula occludens-1 (ZO-1) from tight junctions plays a substantial role in epithelial cell plasticity observed during tumor progression. In vitro, we reported an impact of ZO-1 cyto-nuclear content in modulating the secretion of several pro-inflammatory chemokines. In vivo, we demonstrated that it promotes the recruitment of immune cells in mouse ear sponge assays. Examining lung cancers, we showed that a high density of CD8 cytotoxic T cells and Foxp3 immunosuppressive regulatory T cells in the tumor microenvironment correlated with a cyto-nuclear expression of ZO-1. Taken together, our results support that, by affecting tumor cell secretome, the cyto-nuclear ZO-1 pool may recruit immune cells, which could be permissive for tumor progression.
Collapse
Affiliation(s)
| | - Julien Lesage
- University of Reims Champagne-Ardenne, Inserm UMR-S 1250, SFR CAP-Santé, Reims, France
- Department of Internal Medicine-Medical Oncology, Washington University, St. Louis, MO, United States
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Arnaud Bonnomet
- University of Reims Champagne-Ardenne, Inserm UMR-S 1250, SFR CAP-Santé, Reims, France
- Cellular and Tissue Imaging Platform, University of Reims Champagne-Ardenne, Reims, France
| | - Walter Hunziker
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Agnès Noël
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Valérian Dormoy
- University of Reims Champagne-Ardenne, Inserm UMR-S 1250, SFR CAP-Santé, Reims, France
| | | | - Christine Gilles
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Myriam Polette
- University of Reims Champagne-Ardenne, Inserm UMR-S 1250, SFR CAP-Santé, Reims, France
- Laboratory of Pathology, CHU of Reims, Reims, France
- *Correspondence: Myriam Polette,
| |
Collapse
|
5
|
Duan H, He H, Hu Q, Lin Y, Cao S, Lan X, Li L, Pang D. Comparison of regulatory networks of E74-like factor 1 and cold-shock domain-containing E1 in breast cancer cell lines using ChIP datasets. Exp Ther Med 2020; 20:245. [PMID: 33178343 PMCID: PMC7651883 DOI: 10.3892/etm.2020.9375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 05/01/2020] [Indexed: 12/25/2022] Open
Abstract
In the present study, differences in the expression of target genes between chromatin immunoprecipitation sequencing (ChIP-seq) datasets of breast cancer MCF-7 cells treated with antibodies to E74-like factor 1 (ELF1) and cold-shock domain-containing E1 (CSDE1) were analyzed and gene regulatory networks were established. The datasets were downloaded from the Gene Expression Omnibus (GEO) database. ELF1-associated target genes and CSDE1-associated target genes were analyzed for functional prediction and protein-protein interaction (PPI) networks. The ELF1 ChIP-seq dataset contained 95 ELF1-associated target genes, while the CSDE1 ChIP-seq dataset contained 826 CSDE1-associated target genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis indicated that the ELF1- and CSDE1-associated target genes had different potential functions and signaling pathways. The ELF1-associated target genes were mainly enriched in the GO terms of molecular transducer activity, catalytic activity, cellular processes and response to sensitivity, and in the KEGG pathways of olfactory transduction, the chemokine signaling pathway, carbohydrate digestion and absorption, and starch and sucrose metabolism. The CSDE1-associated target genes were mainly enriched in the GO terms of binding, transcription regulator activity, cellular processes and metabolic processes, and in the KEGG pathways of ribosome, metabolic pathways, endocytosis, oxidative phosphorylation and transcriptional misregulation in cancer. PPI network analysis revealed that the ELF1 regulatory network primarily regulated chemokine-mediated malignant tumor cells, while the CSDE1 regulatory network mainly regulated ribosomes, metabolic pathways and oxidative phosphorylation. Reverse transcription-quantitative PCR indicated that ELF1 overexpression led to significant downregulation of C-X-C motif chemokine-8 and -6 expression levels in MCF-7 cells, while overexpression of CSDE1 significantly induced the mRNA expression of CSDE1-associated target genes, which included mitochondrial ribosomal protein L4, NADH: ubiquinone oxidoreductase subunit B7, small nuclear ribonucleoprotein polypeptide E, ribosomal protein S26 (RPS26), RPS11 and RPS6, in the MCF-7 cells. In breast cancer MCF-7 cells, the target genes and regulatory pathways of ELF1 and CSDE1 were different. Understanding these regulatory pathways may help to develop strategies for personalized breast cancer treatment.
Collapse
Affiliation(s)
- Haibo Duan
- Department of Breast Cancer Medicine, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Huafang He
- Department of Radiotherapy, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Qian Hu
- Department of Breast Cancer Medicine, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Yingxin Lin
- Department of Breast Cancer Medicine, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Shuo Cao
- Department of Breast Cancer Medicine, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Xiaoshan Lan
- Department of Breast Cancer Medicine, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Litao Li
- Department of Breast Cancer Medicine, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Danmei Pang
- Department of Breast Cancer Medicine, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| |
Collapse
|
6
|
Milard M, Penhoat A, Durand A, Buisson C, Loizon E, Meugnier E, Bertrand K, Joffre F, Cheillan D, Garnier L, Viel S, Laugerette F, Michalski MC. Acute effects of milk polar lipids on intestinal tight junction expression: towards an impact of sphingomyelin through the regulation of IL-8 secretion? J Nutr Biochem 2019; 65:128-138. [DOI: 10.1016/j.jnutbio.2018.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/30/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
|
7
|
Rotondi M, Coperchini F, Latrofa F, Chiovato L. Role of Chemokines in Thyroid Cancer Microenvironment: Is CXCL8 the Main Player? Front Endocrinol (Lausanne) 2018; 9:314. [PMID: 29977225 PMCID: PMC6021500 DOI: 10.3389/fendo.2018.00314] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/25/2018] [Indexed: 12/31/2022] Open
Abstract
Tumor-related inflammation does influence the biological behavior of neoplastic cells and ultimately the patient's outcome. With specific regard to thyroid cancer, the issue of tumor-associated inflammation has been extensively studied and recently reviewed. However, the role of chemokines, which play a crucial role in determining the immuno-phenotype of tumor-related inflammation, was not addressed in previous reviews on the topic. Experimental evidence shows that thyroid cancer cells actively secrete a wide spectrum of chemokines and, at least for some of them, solid scientific data support a role for these immune-active molecules in the aggressive behavior of the tumor. Our proposal for a review article on chemokines and thyroid cancer stems from the notion that chemokines, besides having the ability to attract and maintain immune cells at the tumor site, also produce several pro-tumorigenic actions, which include proangiogenetic, cytoproliferative, and pro-metastatic effects. Studies taking into account the role of CCL15, C-X-C motif ligand 12, CXCL16, CXCL1, CCL20, and CCL2 in the context of thyroid cancer will be reviewed with particular emphasis on CXCL8. The reason for focusing on CXCL8 is that this chemokine is the most studied one in human malignancies, displaying multifaceted pro-tumorigenic effects. These include enhancement of tumor cells growth, metastatization, and angiogenesis overall contributing to the progression of several cancers including thyroid cancer. We aim at reviewing current knowledge on the (i) ability of both normal and tumor thyroid cells to secrete CXCL8; (ii) direct/indirect pro-tumorigenic effects of CXCL8 demonstrated by in vitro and in vivo studies specifically performed on thyroid cancer cells; and (iii) pharmacologic strategies proven to be effective for lowering CXCL8 secretion and/or its effects on thyroid cancer cells.
Collapse
Affiliation(s)
- Mario Rotondi
- Unit of Internal Medicine and Endocrinology, ICS Maugeri I.R.C.C.S., Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
| | - Francesca Coperchini
- Unit of Internal Medicine and Endocrinology, ICS Maugeri I.R.C.C.S., Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
| | - Francesco Latrofa
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Chiovato
- Unit of Internal Medicine and Endocrinology, ICS Maugeri I.R.C.C.S., Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
- *Correspondence: Luca Chiovato,
| |
Collapse
|
8
|
Wu J, Zhou XJ, Sun X, Xia TS, Li XX, Shi L, Zhu L, Zhou WB, Wei JF, Ding Q. RBM38 is involved in TGF-β-induced epithelial-to-mesenchymal transition by stabilising zonula occludens-1 mRNA in breast cancer. Br J Cancer 2017; 117:675-684. [PMID: 28683467 PMCID: PMC5572167 DOI: 10.1038/bjc.2017.204] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/31/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The transforming growth factor-β (TGF-β) pathway plays a vital role in driving cancer cell epithelial-mesenchymal transition (EMT). Zonula occludens-1 (ZO-1), which is downregulated in response to TGF-β, is able to control endothelial cell-cell tension, cell migration, and barrier formation. However, the molecular mechanism of how TGF-β regulates ZO-1 expression remains unclear. METHODS Breast cancer cells were treated with TGF-β to induce an EMT progress. Chromatin immunoprecipitation and dual-luciferase reporter assay were performed to investigate direct relationship between Snail and RNA binding motif protein 38 (RBM38). The RNA immunoprecipitation combined with RNA electrophoretic mobility shift assay and dual-luciferase reporter assay were conducted to testify direct relationship between RBM38 and ZO-1. The ZO-1 siRNA was transfected to breast cancer cells that overexpress RBM38 and the control, followed by transwell and Matrigel invasion assays to examine cell migratory and invasive ability. RESULTS Transforming growth factor-β induced a remarkable downregulation of RBM38 in breast cancer that was directly regulated by transcription repressor Snail targeting the E-box elements in promoter region of RBM38 gene. Additionally, RBM38 positively regulated ZO-1 transcript via directly binding to AU/U-rich elements in its mRNA 3'-UTR. Moreover, by magnifying RBM38 expression, cell migration and invasion mediated by knockdown of ZO-1 in breast cancer were reversed. CONCLUSIONS All the results clarified a linear regulation relationship among Snail, RBM38, and ZO-1, implicating RBM38 as a pivotal mediator in TGF-β-induced EMT in breast cancer.
Collapse
Affiliation(s)
- Jing Wu
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Xu-Jie Zhou
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Xi Sun
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Tian-Song Xia
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Xiao-Xia Li
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Liang Shi
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Lei Zhu
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Wen-Bin Zhou
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Ji-Fu Wei
- Research Division of Clinical Pharmacology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
9
|
Stecklein SR, Reddy JP, Wolfe AR, Lopez MS, Fouad TM, Debeb BG, Ueno NT, Brewster AM, Woodward WA. Lack of Breastfeeding History in Parous Women with Inflammatory Breast Cancer Predicts Poor Disease-Free Survival. J Cancer 2017; 8:1726-1732. [PMID: 28819368 PMCID: PMC5556634 DOI: 10.7150/jca.20095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose: Breastfeeding alters the breast microenvironment, and several lines of evidence suggest the breast microenvironment contributes to the clinical phenotype of inflammatory breast cancer. We investigated breastfeeding history as a modifier of locoregional recurrence (LRR), distant metastasis (DM), disease-free survival (DFS), and overall survival (OS) in parous women with inflammatory breast cancer. Methods: Parous women with inflammatory breast cancer were identified from a prospective registry at The University of Texas MD Anderson Cancer Center. We compared patient and tumor characteristics, LRR, DM, DFS, and OS patients with (BF+) and without (BF-) a history of breastfeeding. Results: Eighty-two patients were included. At a median follow-up of 50 months, BF+ patients had significantly lower risk of LRR (9.0% vs. 23.6%; p=0.01), a lower risk of DM (26.8% vs. 53.8%; p=0.008), and better DFS (73.1% vs. 48.1%; p=0.006) than BF- patients. On multivariate analysis, BF+ history was associated with significantly lower risk of DM (hazard ratio 0.38, 95% confidence interval 0.15-0.97; p=0.04) and better DFS (hazard ratio 0.37, 95% confidence interval 0.15-0.93; p=0.04) after adjusting for established predictive and prognostic variables. The prognostic significance of breastfeeding may be most pronounced in women with triple-negative IBC. Conclusion: A lack of breastfeeding history in parous women with inflammatory breast cancer may predict worse prognosis. We speculate that breastfeeding-induced alterations in the breast microenvironment may alter the aggressiveness of inflammatory breast cancer.
Collapse
Affiliation(s)
| | | | | | | | - Tamer M Fouad
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America.,Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic
| | - Abenaa M Brewster
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America.,Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Wendy A Woodward
- Department of Radiation Oncology.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic
| |
Collapse
|
10
|
Suarez‐Carmona M, Lesage J, Cataldo D, Gilles C. EMT and inflammation: inseparable actors of cancer progression. Mol Oncol 2017; 11:805-823. [PMID: 28599100 PMCID: PMC5496491 DOI: 10.1002/1878-0261.12095] [Citation(s) in RCA: 404] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/14/2022] Open
Abstract
Tumors can be depicted as wounds that never heal, and are infiltrated by a large array of inflammatory and immune cells. Tumor-associated chronic inflammation is a hallmark of cancer that fosters progression to a metastatic stage, as has been extensively reviewed lately. Indeed, inflammatory cells persisting in the tumor establish a cross-talk with tumor cells that may result in a phenotype switch into tumor-supporting cells. This has been particularly well described for macrophages and is referred to as tumor-associated 'M2' polarization. Epithelial-to-mesenchymal transition (EMT), the embryonic program that loosens cell-cell adherence complexes and endows cells with enhanced migratory and invasive properties, can be co-opted by cancer cells during metastatic progression. Cancer cells that have undergone EMT are more aggressive, displaying increased invasiveness, stem-like features, and resistance to apoptosis. EMT programs can also stimulate the production of proinflammatory factors by cancer cells. Conversely, inflammation is a potent inducer of EMT in tumors. Therefore, the two phenomena may sustain each other, in an alliance for metastasis. This is the focus of this review, where the interconnections between EMT programs and cellular and molecular actors of inflammation are described. We also recapitulate data linking the EMT/inflammation axis to metastasis.
Collapse
Affiliation(s)
- Meggy Suarez‐Carmona
- National Center for Tumor Diseases (NCT) – University Hospital HeidelbergGermany
| | - Julien Lesage
- Laboratory of Tumor and Development BiologyGIGA‐Cancer University of LiègeBelgium
| | - Didier Cataldo
- Inserm UMR‐S 903SFR CAP‐SantéUniversity of Reims Champagne‐Ardenne (URCA)France
| | - Christine Gilles
- Inserm UMR‐S 903SFR CAP‐SantéUniversity of Reims Champagne‐Ardenne (URCA)France
| |
Collapse
|
11
|
Lesage J, Suarez‐Carmona M, Neyrinck‐Leglantier D, Grelet S, Blacher S, Hunziker W, Birembaut P, Noël A, Nawrocki‐Raby B, Gilles C, Polette M. Zonula occludens‐1/NF‐κB/CXCL8: a new regulatory axis for tumor angiogenesis. FASEB J 2017; 31:1678-1688. [DOI: 10.1096/fj.201600890r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/03/2017] [Indexed: 01/18/2023]
Affiliation(s)
- Julien Lesage
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Meggy Suarez‐Carmona
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Deborah Neyrinck‐Leglantier
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Simon Grelet
- Department of Biochemistry and Molecular BiologyMedical University of South Carolina Charleston South Carolina USA
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Walter Hunziker
- Epithelial Cell Biology LaboratoryInstitute of Molecular and Cell Biology Singapore Singapore
| | - Philippe Birembaut
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
- Laboratory of BiopathologyCentres Hospitaliers Universitaires Reims France
| | - Agnes Noël
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Béatrice Nawrocki‐Raby
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Christine Gilles
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Myriam Polette
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
- Laboratory of BiopathologyCentres Hospitaliers Universitaires Reims France
| |
Collapse
|
12
|
Li XP, Yang XY, Biskup E, Zhou J, Li HL, Wu YF, Chen ML, Xu F. Co-expression of CXCL8 and HIF-1α is associated with metastasis and poor prognosis in hepatocellular carcinoma. Oncotarget 2016; 6:22880-9. [PMID: 26078356 PMCID: PMC4673206 DOI: 10.18632/oncotarget.4412] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/25/2015] [Indexed: 12/24/2022] Open
Abstract
Hypoxia inducible factor-1α (HIF-1α), induces cytokines such as CXCL8 and tumor dissemination, chemo- and radio-resistance. We analyzed correlation between HIF-1α and CXCL8 levels, tumor characteristics and overall survival in 102 hepatocellular carcinoma (HCC) patients. Levels of HIF-1α and CXCL8 were increased in HCC tissues and cell lines. Patients with high levels of HIF-1α and CXCL8 had worse outcome and poorer prognosis than those with lower levels. Co-overexpression of HIF-1α and CXCL8 was an independent negative prognostic factor for overall and disease-free survival. HIF-1α silencing and CXCL8 siRNA decreased migration under hypoxic conditions in vitro. Hypoxia-induced activation of AKT/mTOR/STAT3 pathways was reversed by depletion of CXCL8. We conclude that HIF-1α and CXCL8 induce HCC progression and metastasis, associated with activation of AKT/mTOR/STAT3. Co-expression of HIF-1α and CXCL8 is a prognostic marker and a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Xian-Peng Li
- Division of Gastroenterology and Hepatology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, China
| | - Xiao-Yu Yang
- Division of Special Treatment II, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Ewelina Biskup
- Department of Oncology, University Hospital of Basel, Basel, Switzerland
| | - Jiang Zhou
- Division of Gastroenterology and Hepatology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, China
| | - Hong-Liang Li
- Division of Gastroenterology and Hepatology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, China
| | - Yi-Feng Wu
- Division of Hepatobiliary Surgery, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, China
| | - Ming-Liang Chen
- Division of Hepatobiliary Surgery, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, China
| | - Feng Xu
- Division of Gastroenterology and Hepatology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, China
| |
Collapse
|
13
|
Gonzalez-Mariscal L, Miranda J, Ortega-Olvera JM, Gallego-Gutierrez H, Raya-Sandino A, Vargas-Sierra O. Zonula Occludens Proteins in Cancer. CURRENT PATHOBIOLOGY REPORTS 2016. [DOI: 10.1007/s40139-016-0109-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
14
|
David JM, Dominguez C, Hamilton DH, Palena C. The IL-8/IL-8R Axis: A Double Agent in Tumor Immune Resistance. Vaccines (Basel) 2016; 4:vaccines4030022. [PMID: 27348007 PMCID: PMC5041016 DOI: 10.3390/vaccines4030022] [Citation(s) in RCA: 276] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/31/2016] [Accepted: 06/21/2016] [Indexed: 12/13/2022] Open
Abstract
Interleukin-8 (IL-8, CXCL8) is a pro-inflammatory chemokine produced by various cell types to recruit leukocytes to sites of infection or tissue injury. Acquisition of IL-8 and/or its receptors CXCR1 and CXCR2 are known to be a relatively common occurrence during tumor progression. Emerging research now indicates that paracrine signaling by tumor-derived IL-8 promotes the trafficking of neutrophils and myeloid-derived suppressor cells (MDSCs) into the tumor microenvironment, which have the ability to dampen anti-tumor immune responses. Furthermore, recent studies have also shown that IL-8 produced by the tumor mass can induce tumor cells to undergo the transdifferentiation process epithelial-to-mesenchymal transition (EMT) in which tumor cells shed their epithelial characteristics and acquire mesenchymal characteristics. EMT can increase metastatic dissemination, stemness, and intrinsic resistance, including to killing by cytotoxic immune cells. This review highlights the dual potential roles that the inflammatory cytokine IL-8 plays in promoting tumor resistance by enhancing the immunosuppressive microenvironment and activating EMT, and then discusses the potential for targeting the IL-8/IL-8 receptor axis to combat these various resistance mechanisms.
Collapse
Affiliation(s)
- Justin M David
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Charli Dominguez
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Duane H Hamilton
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
15
|
Kumar S, Chaudhary AK, Kumar R, O'Malley J, Dubrovska A, Wang X, Yadav N, Goodrich DW, Chandra D. Combination therapy induces unfolded protein response and cytoskeletal rearrangement leading to mitochondrial apoptosis in prostate cancer. Mol Oncol 2016; 10:949-65. [PMID: 27106131 DOI: 10.1016/j.molonc.2016.03.007] [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: 01/13/2016] [Revised: 03/13/2016] [Accepted: 03/23/2016] [Indexed: 02/07/2023] Open
Abstract
Development of therapeutic resistance is responsible for most prostate cancer (PCa) related mortality. Resistance has been attributed to an acquired or selected cancer stem cell phenotype. Here we report the histone deacetylase inhibitor apicidin (APC) or ER stressor thapsigargin (TG) potentiate paclitaxel (TXL)-induced apoptosis in PCa cells and limit accumulation of cancer stem cells. TXL-induced responses were modulated in the presence of TG with increased accumulation of cells at G1-phase, rearrangement of the cytoskeleton, and changes in cytokine release. Cytoskeletal rearrangement was associated with modulation of the cytoplasmic and mitochondrial unfolded protein response leading to mitochondrial dysfunction and release of proapoptotic proteins from mitochondria. TXL in combination with APC or TG enhanced caspase activation. Importantly, TXL in combination with TG induced caspase activation and apoptosis in X-ray resistant LNCaP cells. Increased release of transforming growth factor-beta (TGF-β) was observed while phosphorylated β-catenin level was suppressed with TXL combination treatments. This was accompanied by a decrease in the CD44(+)CD133(+) cancer stem cell-like population, suggesting treatment affects cancer stem cell properties. Taken together, combination treatment with TXL and either APC or TG induces efficient apoptosis in both proliferating and cancer stem cells, suggesting this therapeutic combination may overcome drug resistance and recurrence in PCa.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse, Dresden, Germany; German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Xinjiang Wang
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - David W Goodrich
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
16
|
Lee SH, Paek AR, Yoon K, Kim SH, Lee SY, You HJ. Tight junction protein 1 is regulated by transforming growth factor-β and contributes to cell motility in NSCLC cells. BMB Rep 2015; 48:115-20. [PMID: 25027596 PMCID: PMC4352613 DOI: 10.5483/bmbrep.2015.48.2.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Indexed: 01/15/2023] Open
Abstract
Tight junction protein 1 (TJP1), a component of tight junction, has been reported to play a role in protein networks as an adaptor protein, and TJP1 expression is altered during tumor development. Here, we found that TJP1 expression was increased at the RNA and protein levels in TGF-β-stimulated lung cancer cells, A549. SB431542, a type-I TGF-β receptor inhibitor, as well as SB203580, a p38 kinase inhibitor, significantly abrogated the effect of TGF-β on TJP1 expression. Diphenyleneiodonium, an NADPH oxidase inhibitor, also attenuated TJP1 expression in response to TGF-β in lung cancer cells. When TJP1 expression was reduced by shRNA lentiviral particles in A549 cells (A549-sh TJP1), wound healing was much lower than in cells infected with control viral particles. Taken together, these data suggest that TGF-β enhances TJP1 expression, which may play a role beyond structural support in tight junctions during cancer development.
Collapse
Affiliation(s)
- So Hee Lee
- Cancer Cell and Molecular Biology Branch, Div. of Cancer Biology, National Cancer Center, Goyang 410-769; Division of Molecular Life Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - A Rome Paek
- Cancer Cell and Molecular Biology Branch, Div. of Cancer Biology, National Cancer Center, Goyang 410-769, Korea
| | - Kyungsil Yoon
- Lung Cancer Branch, Div. of Translational and Clinical Research I, National Cancer Center, Goyang 410-769, Korea
| | - Seok Hyun Kim
- Cancer Cell and Molecular Biology Branch, Div. of Cancer Biology, National Cancer Center, Goyang 410-769, Korea
| | - Soo Young Lee
- Division of Molecular Life Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Hye Jin You
- Cancer Cell and Molecular Biology Branch, Div. of Cancer Biology, National Cancer Center, Goyang 410-769, Korea
| |
Collapse
|
17
|
Syndecan-1 Acts in Synergy with Tight Junction Through Stat3 Signaling to Maintain Intestinal Mucosal Barrier and Prevent Bacterial Translocation. Inflamm Bowel Dis 2015; 21:1894-907. [PMID: 25970544 DOI: 10.1097/mib.0000000000000421] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Intestinal epithelial tight junction (TJ) is the principal determinant of mucosal permeability, defects of which have been correlated to inflammatory bowel disease. In this study, we investigated whether syndecan-1 (Sdc1), the predominant cell surface heparan sulfate proteoglycan, affects TJ proteins to protect intestinal barrier function. METHODS The role of Sdc1 in barrier function was examined in cultured colonic epithelial cells and dextran sodium sulfate-induced colitis mouse model. Barrier function was determined by transepithelial electrical resistance, bacterial translocation, and FITC-dextran flux. Canonical TJ proteins ZO-1 and occludin were measured by Western blot and immunofluoresence. Role of the Stat3 pathway was detected by Western blot and chromatin immunoprecipitation. RESULTS Overexpressed Sdc1 in Caco-2 cells attenuated transepithelial electrical resistance reduction, prevented bacterial translocation, and repressed FITC-dextran flux, whereas Sdc1 knockdown in HT29 cells resulted in a greater loss of barrier function. Supplementation of exogenous Sdc1 in colitis mice ameliorated the activity of colitis and barrier defect. Mechanistically, Sdc1 significantly modulated expressions of ZO-1 and occludin by activating Stat3, which directly bound to the promoter regions of ZO-1 and occludin. Furthermore, ZO-1 and occludin were found to bind to each other, and their repression could induce Sdc1 upregulation. CONCLUSIONS Sdc1 plays an important role in protecting the intestinal barrier function and preventing bacterial translocation, in synergy with TJ through Stat3 signaling in an Sdc1/Stat3/ZO-1 and occludin feedback loop. Sdc1 participates in the mechanism that is related to intestinal barrier function and colitis and represents a therapeutic target for novel anti-inflammatory bowel disease strategies.
Collapse
|
18
|
Suarez-Carmona M, Bourcy M, Lesage J, Leroi N, Syne L, Blacher S, Hubert P, Erpicum C, Foidart JM, Delvenne P, Birembaut P, Noël A, Polette M, Gilles C. Soluble factors regulated by epithelial-mesenchymal transition mediate tumour angiogenesis and myeloid cell recruitment. J Pathol 2015; 236:491-504. [PMID: 25880038 DOI: 10.1002/path.4546] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 03/18/2015] [Accepted: 04/13/2015] [Indexed: 01/13/2023]
Abstract
Epithelial-mesenchymal transition (EMT) programmes provide cancer cells with invasive and survival capacities that might favour metastatic dissemination. Whilst signalling cascades triggering EMT have been extensively studied, the impact of EMT on the crosstalk between tumour cells and the tumour microenvironment remains elusive. We aimed to identify EMT-regulated soluble factors that facilitate the recruitment of host cells in the tumour. Our findings indicate that EMT phenotypes relate to the induction of a panel of secreted mediators, namely IL-8, IL-6, sICAM-1, PAI-1 and GM-CSF, and implicate the EMT-transcription factor Snail as a regulator of this process. We further show that EMT-derived soluble factors are pro-angiogenic in vivo (in the mouse ear sponge assay), ex vivo (in the rat aortic ring assay) and in vitro (in a chemotaxis assay). Additionally, conditioned medium from EMT-positive cells stimulates the recruitment of myeloid cells. In a bank of 40 triple-negative breast cancers, tumours presenting features of EMT were significantly more angiogenic and infiltrated by a higher quantity of myeloid cells compared to tumours with little or no EMT. Taken together, our results show that EMT programmes trigger the expression of soluble mediators in cancer cells that stimulate angiogenesis and recruit myeloid cells in vivo, which might in turn favour cancer spread.
Collapse
Affiliation(s)
- Meggy Suarez-Carmona
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium.,Laboratory of Experimental Pathology (LEP), GIGA-Cancer, Liège, Belgium
| | - Morgane Bourcy
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Julien Lesage
- INSERM UMR-S 903, Laboratoire Pol Bouin, University of Reims, France
| | - Natacha Leroi
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Laïdya Syne
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Silvia Blacher
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Pascale Hubert
- Laboratory of Experimental Pathology (LEP), GIGA-Cancer, Liège, Belgium
| | - Charlotte Erpicum
- Laboratory of Experimental Pathology (LEP), GIGA-Cancer, Liège, Belgium
| | - Jean-Michel Foidart
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Philippe Delvenne
- Laboratory of Experimental Pathology (LEP), GIGA-Cancer, Liège, Belgium
| | | | - Agnès Noël
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Myriam Polette
- INSERM UMR-S 903, Laboratoire Pol Bouin, University of Reims, France
| | - Christine Gilles
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| |
Collapse
|
19
|
RhoGTPase signalling at epithelial tight junctions: Bridging the GAP between polarity and cancer. Int J Biochem Cell Biol 2015; 64:120-5. [PMID: 25757376 PMCID: PMC4503795 DOI: 10.1016/j.biocel.2015.02.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/17/2015] [Accepted: 02/26/2015] [Indexed: 01/19/2023]
Abstract
The establishment and maintenance of epithelial polarity must be correctly controlled for normal development and homeostasis. Tight junctions (TJ) in vertebrates define apical and basolateral membrane domains in polarized epithelia via bi-directional, complex signalling pathways between TJ themselves and the cytoskeleton they are associated with. RhoGTPases are central to these processes and evidence suggests that their regulation is coordinated by interactions between GEFs and GAPs with junctional, cytoplasmic adapter proteins. In this InFocus review we determine that the expression, localization or stability of a variety of these adaptor proteins is altered in various cancers, potentially representing an important mechanistic link between loss of polarity and cancer. We focus here, on two well characterized RhoGTPases Cdc42 and RhoA who's GEFs and GAPs are predominantly localized to TJ via cytoplasmic adaptor proteins.
Collapse
|
20
|
Han J, Bae SY, Oh SJ, Lee J, Lee JH, Lee HC, Lee SK, Kil WH, Kim SW, Nam SJ, Kim S, Lee JE. Zerumbone suppresses IL-1β-induced cell migration and invasion by inhibiting IL-8 and MMP-3 expression in human triple-negative breast cancer cells. Phytother Res 2014; 28:1654-60. [PMID: 24890258 DOI: 10.1002/ptr.5178] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/01/2014] [Indexed: 12/19/2022]
Abstract
Inflammation is a key regulatory process in cancer development. Prolonged exposure of breast tumor cells to inflammatory cytokines leads to epithelial-mesenchymal transition, which is the principal mechanism involved in metastasis and tumor invasion. Interleukin (IL)-1β is a major inflammatory cytokine in a variety of tumors. To date, the regulatory mechanism of IL-1β-induced cell migration and invasion has not been fully elucidated. Here, we investigated the effect of zerumbone (ZER) on IL-1β-induced cell migration and invasion in breast cancer cells. The levels of IL-8 and matrix metalloproteinase (MMP)-3 mRNA were analyzed by real-time polymerase chain reaction. The levels of secreted IL-8 and MMP-3 protein were analyzed by enzyme-linked immunosorbent assay and western blot analysis, respectively. Cell invasion and migration was detected by Boyden chamber assay. The levels of IL-8 and MMP-3 expression were significantly increased by IL-1β treatment in Hs578T and MDA-MB231 cells. On the other hand, IL-1β-induced IL-8 and MMP-3 expression was decreased by ZER. Finally, IL-1β-induced cell migration and invasion were decreased by ZER in Hs578T and MDA-MB231 cells. ZER suppresses IL-1β-induced cell migration and invasion by inhibiting IL-8 expression and MMP-3 expression in TNBC cells. ZER could be a promising therapeutic drug for treatment of triple-negative breast cancer patients.
Collapse
Affiliation(s)
- Jeonghun Han
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul, Korea; Department of Life Science, Hanyang University, Seoul, 133-791, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
TRAF4 is a novel phosphoinositide-binding protein modulating tight junctions and favoring cell migration. PLoS Biol 2013; 11:e1001726. [PMID: 24311986 PMCID: PMC3848981 DOI: 10.1371/journal.pbio.1001726] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 10/23/2013] [Indexed: 12/26/2022] Open
Abstract
The cancer-associated TRAF4 protein has a TRAF domain that is a bona fide phosphoinositide-binding domain and involved in the modulation of tight junctions and cell migration. Tumor necrosis factor (TNF) receptor-associated factor 4 (TRAF4) is frequently overexpressed in carcinomas, suggesting a specific role in cancer. Although TRAF4 protein is predominantly found at tight junctions (TJs) in normal mammary epithelial cells (MECs), it accumulates in the cytoplasm of malignant MECs. How TRAF4 is recruited and functions at TJs is unclear. Here we show that TRAF4 possesses a novel phosphoinositide (PIP)-binding domain crucial for its recruitment to TJs. Of interest, this property is shared by the other members of the TRAF protein family. Indeed, the TRAF domain of all TRAF proteins (TRAF1 to TRAF6) is a bona fide PIP-binding domain. Molecular and structural analyses revealed that the TRAF domain of TRAF4 exists as a trimer that binds up to three lipids using basic residues exposed at its surface. Cellular studies indicated that TRAF4 acts as a negative regulator of TJ and increases cell migration. These functions are dependent from its ability to interact with PIPs. Our results suggest that TRAF4 overexpression might contribute to breast cancer progression by destabilizing TJs and favoring cell migration. Tumor necrosis factor (TNF) receptor-associated factor 4, also known as TRAF4, is an unusual member of the TRAF protein family. While TRAFs are primarily known as regulators of inflammation, antiviral responses, and apoptosis, research on TRAF4 has identified its involvement in development and cancer. Importantly TRAF4 overexpression has been associated with a poor prognosis in breast cancers. TRAF4 has multiple subcellular localizations: to the plasma membrane in tight junctions (TJs), cytoplasmic and nuclear. The recruitment mechanisms and the functional impact of these distinct localizations are not completely understood. Here we investigate how TRAF4 is recruited to TJs and its involvement in cell–cell contacts in mammary epithelial cells (MECs). We show that the TRAF domain of all TRAFs contains a lipid binding module, and that TRAF4 uses this domain to form a trimeric complex that associates with phosphoinositides at the plasma membrane. Moreover this interaction is necessary for its recruitment to TJs. Additionally, we show that through its interaction with lipids TRAF4 delays TJ assembly and increases cell migration. We propose that TRAF4 has an important function in cancer progression by destabilizing TJs and favoring cell migration.
Collapse
|
22
|
The Chemokine CXCL8 in Carcinogenesis and Drug Response. ISRN ONCOLOGY 2013; 2013:859154. [PMID: 24224100 PMCID: PMC3810054 DOI: 10.1155/2013/859154] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/08/2013] [Indexed: 02/08/2023]
Abstract
Although the functions of chemokines in the regulation of immune processes have been studied in some detail, the role of these biomolecules in cancer is not fully understood. Chemokines mediate migration of immune cells and other functions related to immunity. They are also involved in oncogenesis and in tumor progression, invasion, and metastasis through mechanisms similar to their roles in immune functions. Various chemokines also promote cell proliferation and resistance to apoptosis of stressed cells. Consequently, chemokines and their receptors present potential therapeutic targets for anticancer drugs. The chemokine CXCL8, also known as interleukin-8 (IL8), is a proinflammatory molecule that has functions within the tumor microenvironment. Due to its potent angiogenic effects and the activity of the chemokine and its receptors in the promotion of invasion and metastasis, CXCL8 and its receptors are now considered as attractive targets for cancer therapy. This review relates the current understanding of the regulation, signaling, and functions of CXCL8 that contribute to tumor growth and metastasis, and of its role in drug response.
Collapse
|
23
|
Zhu Q, Han X, Peng J, Qin H, Wang Y. The role of CXC chemokines and their receptors in the progression and treatment of tumors. J Mol Histol 2012; 43:699-713. [PMID: 22752457 DOI: 10.1007/s10735-012-9435-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/21/2012] [Indexed: 12/25/2022]
Abstract
Chemokines are a class of functional chemotactic peptides that contribute to a number of tumor-related processes. They are functionally defined as soluble factors that are able to control the directional migration of leukocytes, in particular, during infection and inflammation. It appears, however, that the biological effects mediated by chemokines are far more complex, and virtually all cells, including many tumor cell types, can express chemokines and chemokine receptors. A growing body of evidence indicates that they also contribute to a number of tumor-related processes, such as tumor cell growth, angiogenesis/angiostasis, local invasion, and mediate organ-specific metastases of cancer. The CXC chemokine class is a subfamily of a large family of chemokines. During the occurrence and development of tumor cells, this chemokine class is often accompanied by a series of molecular and biological changes. The CXC chemokine subfamily is closely related to the body's immune response to tumors and biological behaviors of tumors. In this paper, CXC chemokines and their role in the progression and treatment of tumors will be reviewed.
Collapse
Affiliation(s)
- Qingchao Zhu
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, 200233, People's Republic of China.
| | | | | | | | | |
Collapse
|