1
|
Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024; 45:1337-1348. [PMID: 38351317 PMCID: PMC11192764 DOI: 10.1038/s41401-024-01235-6] [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: 10/05/2023] [Accepted: 01/28/2024] [Indexed: 02/19/2024] Open
Abstract
Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-β signaling pathways leads to pathological conditions. TGF-β signaling is regulated at different levels along the pathways and begins with the liberation of TGF-β ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-β isoforms, enabling precise control of TGF-β signals. In addition, the cell surface compartments used to release active TGF-β are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - Ying-Nan Cao
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia.
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
| |
Collapse
|
2
|
Nishimura H, Jin D, Kinoshita I, Taniuchi M, Higashino M, Terada T, Takai S, Kawata R. Increased Chymase-Positive Mast Cells in High-Grade Mucoepidermoid Carcinoma of the Parotid Gland. Int J Mol Sci 2023; 24:ijms24098267. [PMID: 37175975 PMCID: PMC10179695 DOI: 10.3390/ijms24098267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
It has long been known that high-grade mucoepidermoid carcinoma (MEC) has a poor prognosis, but the detailed molecular and biological mechanisms underlying this are not fully understood. In the present study, the pattern of chymase-positive mast cells, as well as chymase gene expression, in high-grade MEC was compared to that of low-grade and intermediate-grade MEC by using 44 resected tumor samples of MEC of the parotid gland. Chymase expression, as well as chymase-positive mast cells, was found to be markedly increased in high-grade MEC. Significant increases in PCNA-positive cells and VEGF gene expression, as well as lymphangiogenesis, were also confirmed in high-grade MEC. Chymase substrates, such as the latent transforming growth factor-beta (TGF-β) 1 and pro-matrix metalloproteinase (MMP)-9, were also detected immunohistologically in high-grade MEC. These findings suggested that the increased chymase activity may increase proliferative activity, as well as metastasis in the malignant condition, and the inhibition of chymase may be a strategy to improve the poor prognosis of high-grade MEC of the parotid gland.
Collapse
Affiliation(s)
- Hiromi Nishimura
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki-City 569-8686, Osaka, Japan
| | - Denan Jin
- Department of Innovative Medicine, Graduate School of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki-City 569-8686, Osaka, Japan
| | - Ichita Kinoshita
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki-City 569-8686, Osaka, Japan
| | - Masataka Taniuchi
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki-City 569-8686, Osaka, Japan
| | - Masaaki Higashino
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki-City 569-8686, Osaka, Japan
| | - Tetsuya Terada
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki-City 569-8686, Osaka, Japan
| | - Shinji Takai
- Department of Innovative Medicine, Graduate School of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki-City 569-8686, Osaka, Japan
| | - Ryo Kawata
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki-City 569-8686, Osaka, Japan
| |
Collapse
|
3
|
He L, Kang Q, Chan KI, Zhang Y, Zhong Z, Tan W. The immunomodulatory role of matrix metalloproteinases in colitis-associated cancer. Front Immunol 2023; 13:1093990. [PMID: 36776395 PMCID: PMC9910179 DOI: 10.3389/fimmu.2022.1093990] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 01/22/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are an important class of enzymes in the body that function through the extracellular matrix (ECM). They are involved in diverse pathophysiological processes, such as tumor invasion and metastasis, cardiovascular diseases, arthritis, periodontal disease, osteogenesis imperfecta, and diseases of the central nervous system. MMPs participate in the occurrence and development of numerous cancers and are closely related to immunity. In the present study, we review the immunomodulatory role of MMPs in colitis-associated cancer (CAC) and discuss relevant clinical applications. We analyze more than 300 pharmacological studies retrieved from PubMed and the Web of Science, related to MMPs, cancer, colitis, CAC, and immunomodulation. Key MMPs that interfere with pathological processes in CAC such as MMP-2, MMP-3, MMP-7, MMP-9, MMP-10, MMP-12, and MMP-13, as well as their corresponding mechanisms are elaborated. MMPs are involved in cell proliferation, cell differentiation, angiogenesis, ECM remodeling, and the inflammatory response in CAC. They also affect the immune system by modulating differentiation and immune activity of immune cells, recruitment of macrophages, and recruitment of neutrophils. Herein we describe the immunomodulatory role of MMPs in CAC to facilitate treatment of this special type of colon cancer, which is preceded by detectable inflammatory bowel disease in clinical populations.
Collapse
Affiliation(s)
- Luying He
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Ka Iong Chan
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China,*Correspondence: Zhangfeng Zhong, ; Wen Tan,
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, China,*Correspondence: Zhangfeng Zhong, ; Wen Tan,
| |
Collapse
|
4
|
Wang Q, Zhao B, Li J, Zhao J, Wang C, Li Q, Yang W, Xu L, Gong Y. Qilian Formula Inhibits Tumor Cell Growth in a Bone Metastasis Model of Lung Cancer. Integr Cancer Ther 2023; 22:15347354231217274. [PMID: 38130184 DOI: 10.1177/15347354231217274] [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: 12/23/2023] Open
Abstract
BACKGROUND Bone metastasis is frequently common in advanced lung cancer with the major issue of a pathological fracture. Previous studies suggested that Astragalus membranaceus (Qi) and Ampelopsis japonica (Lian), which are used as folk medicine in China, have potential effects on inhibiting tumor growth and protecting bones, respectively. In this study, an experiment on the inhibitory effect of the Qilian formula (AAF) in vivo was designed to examine tumor growth in bone and osteoclast formation. MATERIALS AND METHODS The bone metastasis xenograft models were established by implanting NCI-H460-luc2 lung cancer cells into the right tibiae bones of mice. After confirming the model's viability through optical imaging 7 days post-implantation, 2 groups, namely the AAF group and the control group, were administered 0.3 mL of AAF extract (9 g/kg/day) or normal saline via intragastric delivery for a duration of 4 weeks. Throughout the study, we longitudinally assessed tumor burden, bone destruction, and weight-bearing capacity in vivo using reporter gene bioluminescence imaging (BLI), micro-CT, and dynamic weight-bearing (DWB) tests. Mechanistic insights were gained through Hematoxylin-eosin (H&E) staining, immunohistochemical (IHC) analysis, western blotting, and flow cytometry. RESULTS Qilian formula produced significant inhibition to the progress of bone destruction and tumor burden in the right tibiae bone in the treatment group. It was further evidenced by molecular imaging in vivo via small animal micro-CT and BLI with parametric quantification, characterizing significantly lower uptake of BLI signal in the treated tumor lesions and improving the pathological changes in the microstructure of bone. Furthermore, DWB tests revealed that Qilian formula treatment significantly maintained the weight-bearing capacity. According to immunohistochemical analysis, the effect of the Qilian formula appeared to involve the suppression of osteoclast formation by lower expression of the tartrate-resistant acid phosphatase. Cell apoptosis and death induction were evidenced by a higher percentage of Bal2、BAX and caspase 3 expressions of Qilian formula-treated tumor tissues. CONCLUSIONS Our study demonstrated a significant inhibitory effect of the Qilian formula on the progression of osteolytic invasion in vivo by suppressing osteoclastogenesis and promoting apoptotic cell death.
Collapse
Affiliation(s)
- Qin Wang
- Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bei Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiaqi Li
- Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingwen Zhao
- Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengyan Wang
- Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Quanyao Li
- Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenxiao Yang
- Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Xu
- Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Tumor Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yabin Gong
- Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Riggers DS, Rosati M, Köhler C, Matiasek K, Loderstedt S. A case of extraosseous intradural osteosarcoma of the spine in a dog. VETERINARY RECORD CASE REPORTS 2022. [DOI: 10.1002/vrc2.470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Denise Sarina Riggers
- Small Animal Department, Faculty of Veterinary Medicine University of Leipzig Leipzig Germany
| | - Marco Rosati
- Section of Clinical & Comparative Neuropathology, Centre for Clinical Veterinary Medicine Ludwig‐Maximilians‐Universität München Munich Germany
| | - Claudia Köhler
- Small Animal Department, Faculty of Veterinary Medicine University of Leipzig Leipzig Germany
| | - Kaspar Matiasek
- Section of Clinical & Comparative Neuropathology, Centre for Clinical Veterinary Medicine Ludwig‐Maximilians‐Universität München Munich Germany
| | - Shenja Loderstedt
- Small Animal Department, Faculty of Veterinary Medicine University of Leipzig Leipzig Germany
| |
Collapse
|
6
|
The Synergistic Cooperation between TGF-β and Hypoxia in Cancer and Fibrosis. Biomolecules 2022; 12:biom12050635. [PMID: 35625561 PMCID: PMC9138354 DOI: 10.3390/biom12050635] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/10/2022] [Accepted: 04/14/2022] [Indexed: 12/24/2022] Open
Abstract
Transforming growth factor β (TGF-β) is a multifunctional cytokine regulating homeostasis and immune responses in adult animals and humans. Aberrant and overactive TGF-β signaling promotes cancer initiation and fibrosis through epithelial–mesenchymal transition (EMT), as well as the invasion and metastatic growth of cancer cells. TGF-β is a key factor that is active during hypoxic conditions in cancer and is thereby capable of contributing to angiogenesis in various types of cancer. Another potent role of TGF-β is suppressing immune responses in cancer patients. The strong tumor-promoting effects of TGF-β and its profibrotic effects make it a focus for the development of novel therapeutic strategies against cancer and fibrosis as well as an attractive drug target in combination with immune regulatory checkpoint inhibitors. TGF-β belongs to a family of cytokines that exert their function through signaling via serine/threonine kinase transmembrane receptors to intracellular Smad proteins via the canonical pathway and in combination with co-regulators such as the adaptor protein and E3 ubiquitin ligases TRAF4 and TRAF6 to promote non-canonical pathways. Finally, the outcome of gene transcription initiated by TGF-β is context-dependent and controlled by signals exerted by other growth factors such as EGF and Wnt. Here, we discuss the synergistic cooperation between TGF-β and hypoxia in development, fibrosis and cancer.
Collapse
|
7
|
Wang WK, Wang B, Cao XH, Liu YS. Spironolactone alleviates myocardial fibrosis via inhibition of Ets‑1 in mice with experimental autoimmune myocarditis. Exp Ther Med 2022; 23:369. [PMID: 35495592 PMCID: PMC9019666 DOI: 10.3892/etm.2022.11296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Spironolactone improves cardiac structure, function and prognosis in patients with heart failure and delays the progression of cardiac fibrosis. However, the exact underlying mechanism of this process remains to be elucidated. The present study therefore aimed to explore the protective effect and underlying mechanism of the aldosterone receptor antagonist, spironolactone, on myocardial fibrosis in mice with experimental autoimmune myocarditis (EAM). The EAM model was induced in BALB/c mice via immunization with murine cardiac α-myosin heavy chain sequence polypeptides. The cardiac function of the mice was assessed using echocardiography and the levels of inflammatory cytokines were quantified using ELISA. E26 transformation-specific sequence-1 (Ets-1) expression was knocked down using lentivirus-mediated small interference RNA. Total collagen deposition was assessed using Masson's trichrome and Ets-1, TGF-β1, Smad2/3, collagen I and III protein expression levels were detected using immunohistochemistry and western blotting. MMP-2 and MMP-9 mRNA expression levels and activity was determined using reverse transcription-quantitative PCR and gelatin zymography, respectively. The results of the present study demonstrated that spironolactone significantly improved myocardium hypertrophy, diastolic cardiac function and decreased myocardial inflammation and collagen deposition induced by EAM. Spironolactone treatment significantly inhibited Ets-1 and smad2/3 phosphorylation. In addition, inhibition of Ets-1 reduced the expression and activity of MMP-2 and MMP-9 and decreased cardiac fibrosis in EAM mice. The results indicated that the improvement of myocardial fibrosis by spironolactone may be associated with the TGF-β1/Smad-2/3/Ets-1 signaling pathway in EAM mice.
Collapse
Affiliation(s)
- Wen-Ke Wang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, P.R. China
| | - Ben Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xue-Hu Cao
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yu-Sheng Liu
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
8
|
Zhang B, Li Y, Wu Q, Xie L, Barwick B, Fu C, Li X, Wu D, Xia S, Chen J, Qian WP, Yang L, Osunkoya AO, Boise L, Vertino PM, Zhao Y, Li M, Chen HR, Kowalski J, Kucuk O, Zhou W, Dong JT. Acetylation of KLF5 maintains EMT and tumorigenicity to cause chemoresistant bone metastasis in prostate cancer. Nat Commun 2021; 12:1714. [PMID: 33731701 PMCID: PMC7969754 DOI: 10.1038/s41467-021-21976-w] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/18/2021] [Indexed: 12/25/2022] Open
Abstract
Advanced prostate cancer (PCa) often develops bone metastasis, for which therapies are very limited and the underlying mechanisms are poorly understood. We report that bone-borne TGF-β induces the acetylation of transcription factor KLF5 in PCa bone metastases, and acetylated KLF5 (Ac-KLF5) causes osteoclastogenesis and bone metastatic lesions by activating CXCR4, which leads to IL-11 secretion, and stimulating SHH/IL-6 paracrine signaling. While essential for maintaining the mesenchymal phenotype and tumorigenicity, Ac-KLF5 also causes resistance to docetaxel in tumors and bone metastases, which is overcome by targeting CXCR4 with FDA-approved plerixafor. Establishing a mechanism for bone metastasis and chemoresistance in PCa, these findings provide a rationale for treating chemoresistant bone metastasis of PCa with inhibitors of Ac-KLF5/CXCR4 signaling.
Collapse
Affiliation(s)
- Baotong Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Yixiang Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Qiao Wu
- Department of Genetics and Cell Biology, Nankai University College of Life Sciences, Tianjin, China
| | - Lin Xie
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Third Affiliated Hospital of Kunming Medical University, Cancer Hospital of Yunnan Province, Kunming, China
| | - Benjamin Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Changying Fu
- Department of Genetics and Cell Biology, Nankai University College of Life Sciences, Tianjin, China
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology School of Medicine, Shenzhen, China
| | - Xin Li
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Daqing Wu
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Siyuan Xia
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jing Chen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Wei Ping Qian
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Lily Yang
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Adeboye O Osunkoya
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Pathology and Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lawrence Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Paula M Vertino
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yichao Zhao
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Menglin Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Hsiao-Rong Chen
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jeanne Kowalski
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Wei Zhou
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jin-Tang Dong
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology School of Medicine, Shenzhen, China.
| |
Collapse
|
9
|
Zhao H, Wei J, Sun J. Roles of TGF-β signaling pathway in tumor microenvirionment and cancer therapy. Int Immunopharmacol 2020; 89:107101. [PMID: 33099067 DOI: 10.1016/j.intimp.2020.107101] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/10/2020] [Accepted: 10/10/2020] [Indexed: 12/15/2022]
Abstract
Transforming growth factor β (TGF- β) signaling pathway has pleiotropic effects on cell proliferation, differentiation, adhesion, senescence, and apoptosis. TGF-β can be widely produced by various immune or non-immune cells and regulate cell behaviors through autocrine and paracrine. It plays essential roles in biological processes including embryological development, immune response, and tumor progression. Few cell signalings can contribute to so many pleiotropic functions as the TGF- β signaling pathway in mammals. The significant function of TGF-β signaling in tumor progression and evasion leading it to draw great attention in scientific and clinical research. Understanding the mechanism of TGF- β signaling provides us with chances to potentiate the effectiveness and selectivity of this therapeutic method. Herein, we review the molecular and cellular mechanisms of TGF-β signaling in carcinomas and tumor microenvironment. Then, we enumerate main achievements of TGF-β blockades used or being evaluated in cancer therapy, providing us opportunities to improve therapeutical approaches in the tumor which thrive in a TGF-β-rich environment.
Collapse
Affiliation(s)
- Haodi Zhao
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 30072, PR China
| | - Jing Wei
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Jian Sun
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 30072, PR China; Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| |
Collapse
|
10
|
Das V, Bhattacharya S, Chikkaputtaiah C, Hazra S, Pal M. The basics of epithelial-mesenchymal transition (EMT): A study from a structure, dynamics, and functional perspective. J Cell Physiol 2019; 234:14535-14555. [PMID: 30723913 DOI: 10.1002/jcp.28160] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a key step in transdifferentiation process in solid cancer development. Forthcoming evidence suggest that the stratified program transforms polarized, immotile epithelial cells to migratory mesenchymal cells associated with enhancement of breast cancer stemness, metastasis, and drug resistance. It involves primarily several signaling pathways, such as transforming growth factor-β (TGF-β), cadherin, notch, plasminogen activator protein inhibitor, urokinase plasminogen activator, and WNT/beta catenin pathways. However, current understanding on the crosstalk of multisignaling pathways and assemblies of key transcription factors remain to be explored. In this review, we focus on the crosstalk of signal transduction pathways linked to the current therapeutic and drug development strategies. We have also performed the computational modeling on indepth the structure and conformational dynamic studies of regulatory proteins and analyze molecular interactions with their associate factors to understand the complicated process of EMT in breast cancer progression and metastasis. Electrostatic potential surfaces have been analyzed that help in optimization of electrostatic interactions between the protein and its ligand. Therefore, understanding the biological implications underlying the EMT process through molecular biology with biocomputation and structural biology approaches will enable the development of new therapeutic strategies to sensitize tumors to conventional therapy and suppress their metastatic phenotype.
Collapse
Affiliation(s)
- Vishal Das
- Biological Sciences and Technology Division (Biotechnology Group), CSIR-North East Institute of Science and Technology, Academy of Scientific and Innovative Research, Jorhat, Assam, India
| | - Sourya Bhattacharya
- Department of Biotechnology, Centre for Nanotechnology, Indian Institute of Technology Roorkee (IITR), Roorkee, Uttarakhand, India
| | - Channakeshavaiah Chikkaputtaiah
- Biological Sciences and Technology Division (Biotechnology Group), CSIR-North East Institute of Science and Technology, Academy of Scientific and Innovative Research, Jorhat, Assam, India
| | - Saugata Hazra
- Department of Biotechnology, Centre for Nanotechnology, Indian Institute of Technology Roorkee (IITR), Roorkee, Uttarakhand, India
| | - Mintu Pal
- Biological Sciences and Technology Division (Biotechnology Group), CSIR-North East Institute of Science and Technology, Academy of Scientific and Innovative Research, Jorhat, Assam, India
| |
Collapse
|
11
|
Brook N, Brook E, Dharmarajan A, Dass CR, Chan A. Breast cancer bone metastases: pathogenesis and therapeutic targets. Int J Biochem Cell Biol 2018; 96:63-78. [DOI: 10.1016/j.biocel.2018.01.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/31/2017] [Accepted: 01/04/2018] [Indexed: 01/03/2023]
|
12
|
Hayashi N, Iwamoto T, Qi Y, Niikura N, Santarpia L, Yamauchi H, Nakamura S, Hortobagyi GN, Pusztai L, Symmans WF, Ueno NT. Bone metastasis-related signaling pathways in breast cancers stratified by estrogen receptor status. J Cancer 2017; 8:1045-1052. [PMID: 28529618 PMCID: PMC5436258 DOI: 10.7150/jca.13690] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background: Breast cancer bone metastasis (BCBM)-specific genes have been reported without considering biological differences based on estrogen receptor (ER) status. The aims of this study were to identify BCBM-specific genes using our patient dataset and validate previously reported BCBM-specific genes, and to determine whether ER-status-related biological differences matter in identification of BCBM-specific genes. Methods: We used Affymetrix GeneChips to analyze 365 primary human epidermal growth factor receptor 2 (HER2)-negative invasive breast cancer specimens. Genes that were differentially expressed between patients who developed bone metastasis and those who developed non-bone metastasis were identified using Cox proportional hazards model, and differential expression of gene sets was assessed using gene set analysis. We performed gene set analysis to determine whether biological function associated with bone metastasis were different by ER status using 2,246 functionally annotated gene sets assembled from Gene Ontology data base. Results: Among 16,712 probe sets, 592 were overexpressed in the bone metastasis cohort compared to the non-bone-metastasis cohort (false discovery rate ≤ 0.05). However, no BCBM-specific genes met our significance tests when the cancers were stratified by ER status. In ER-positive and ER-negative breast cancers, 151 and 125 gene sets, respectively, were overexpressed for BCBM and the majority of BCBM-related pathways were different. Of significant gene sets, only 13 gene sets were overlapped between ER-positive and -negative cohorts. Conclusion: ER-positive and ER-negative breast cancers have different biological pathways in BCBM development. We have yet to explore BCBM-related biomarkers and targets considering the biological features associated with BCBM depending on the ER status.
Collapse
Affiliation(s)
- Naoki Hayashi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Takayuki Iwamoto
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuan Qi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoki Niikura
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Libero Santarpia
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hideko Yamauchi
- Department of Breast Surgical Oncology, St. Luke's International Hospital, Tokyo, Japan
| | - Seigo Nakamura
- Department of Surgery, Division of Breast Surgical Oncology, Showa University School of Medicine, Tokyo, Japan
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lajos Pusztai
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W Fraser Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
13
|
Yang W, Wang JG, Xu J, Zhou D, Ren K, Hou C, Chen L, Liu X. HCRP1 inhibits TGF-β induced epithelial-mesenchymal transition in hepatocellular carcinoma. Int J Oncol 2017; 50:1233-1240. [DOI: 10.3892/ijo.2017.3903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 02/07/2017] [Indexed: 11/06/2022] Open
|
14
|
Saed GM, Zhang W, Diamond MP. Effect of Hypoxia on Stimulatory Effect of TGF-β1 on MMP-2 and MMP-9 Activities in Mouse Fibroblasts. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155760000700606] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
| | | | - Michael P. Diamond
- Department of obstetrics and Gynecology, Wayne State University, Detroit, Michigan; Department of Obstetrics and Gynecology, Wayne State Medical School, Wayne State University, 275 E. Hancock, Detroit, MI 48201
| |
Collapse
|
15
|
RAGE and TGF-β1 Cross-Talk Regulate Extracellular Matrix Turnover and Cytokine Synthesis in AGEs Exposed Fibroblast Cells. PLoS One 2016; 11:e0152376. [PMID: 27015414 PMCID: PMC4807770 DOI: 10.1371/journal.pone.0152376] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 01/11/2023] Open
Abstract
AGEs accumulation in the skin affects extracellular matrix (ECM) turnover and triggers diabetes associated skin conditions and accelerated skin aging. The receptor of AGEs (RAGE) has an essential contribution to cellular dysfunction driven by chronic inflammatory responses while TGF-β1 is critical in both dermal homeostasis and inflammation. We investigated the contribution of RAGE and TGF-β1 to the modulation of inflammatory response and ECM turnover in AGEs milieu, using a normal fibroblast cell line. RAGE, TGF-β1, collagen I and III gene and protein expression were upregulated after exposure to AGEs-BSA, and MMP-2 was activated. AGEs-RAGE was pivotal in NF-κB dependent collagen I expression and joined with TGF-β1 to stimulate collagen III expression, probably via ERK1/2 signaling. AGEs-RAGE axis induced upregulation of TGF-β1, TNF-α and IL-8 cytokines. TNF-α and IL-8 were subjected to TGF-β1 negative regulation. RAGE’s proinflammatory signaling also antagonized AGEs-TGF-β1 induced fibroblast contraction, suggesting the existence of an inhibitory cross-talk mechanism between TGF-β1 and RAGE signaling. RAGE and TGF-β1 stimulated anti-inflammatory cytokines IL-2 and IL-4 expression. GM-CSF and IL-6 expression appeared to be dependent only on TGF-β1 signaling. Our data also indicated that IFN-γ upregulated in AGEs-BSA milieu in a RAGE and TGF-β1 independent mechanism. Our findings raise the possibility that RAGE and TGF-β1 are both involved in fibrosis development in a complex cross-talk mechanism, while also acting on their own individual targets. This study contributes to the understanding of impaired wound healing associated with diabetes complications.
Collapse
|
16
|
Yang JS, Lin CW, Su SC, Yang SF. Pharmacodynamic considerations in the use of matrix metalloproteinase inhibitors in cancer treatment. Expert Opin Drug Metab Toxicol 2016; 12:191-200. [PMID: 26852787 DOI: 10.1517/17425255.2016.1131820] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Matrix metalloproteinases (MMPs) are classified in the family of zinc-dependent endopeptidases, which can degrade various components of an extracellular matrix and a basement membrane. Studies have demonstrated that MMPs relate to the development of malignant tumors and induce angiogenesis, resulting in the invasion and metastasis of tumor cells. MMPs are highly expressed in malignant tumors and are related to cancer patients' malignant phenotype and poor prognosis. Therefore, blocking the expression or activity of MMPs may be a promising strategy for cancer treatment. AREAS COVERED This study aimed to explain the MMP structure, regulatory mechanism, and carcinogenic effect; investigate the matrix metalloproteinase-inhibitors (MMPIs) that are currently used in clinical trials for cancer treatment; and summarize the trial results. EXPERT OPINION Currently, the results of clinical trials that have used MMPIs as anticancer agents are unsatisfactory. However, MMPs remain an attractive target for cancer treatment. For example, development of the specific peptide or antibodies in targeting the hemopexin domain of MMP-2 may be a new therapeutic direction. The design and development of MMPIs that have selectivity will be the primary focus in future studies.
Collapse
Affiliation(s)
- Jia-Sin Yang
- a Department of Medical Research , Chung Shan Medical University Hospital , Taichung , Taiwan.,b Institute of Medicine , Chung Shan Medical University , Taichung , Taiwan
| | - Chiao-Wen Lin
- c Institute of Oral Sciences , Chung Shan Medical University , Taichung , Taiwan.,d Department of Dentistry , Chung Shan Medical University Hospital , Taichung , Taiwan
| | - Shih-Chi Su
- e Whole-Genome Research Core Laboratory of Human Diseases , Chang Gung Memorial Hospital , Keelung , Taiwan.,f Department of Dermatology, Drug Hypersensitivity Clinical and Research Center , Chang Gung Memorial Hospitals , Linkou , Taiwan
| | - Shun-Fa Yang
- a Department of Medical Research , Chung Shan Medical University Hospital , Taichung , Taiwan.,b Institute of Medicine , Chung Shan Medical University , Taichung , Taiwan
| |
Collapse
|
17
|
Abstract
The extracellular matrix (ECM) is an essential component of the human body that is responsible for the proper function of various organs. Changes in the ECM have been implicated in the pathogenesis of several cardiovascular conditions including atherosclerosis, restenosis, and heart failure. Matrix components, such as collagens and noncollagenous proteins, influence the function and activity of vascular cells, particularly vascular smooth muscle cells and macrophages. Matrix proteins have been shown to be implicated in the development of atherosclerotic complications, such as plaque rupture, aneurysm formation, and calcification. ECM proteins control ECM remodeling through feedback signaling to matrix metalloproteinases (MMPs), which are the key players of ECM remodeling in both normal and pathological conditions. The production of MMPs is closely related to the development of an inflammatory response and is subjected to significant changes at different stages of atherosclerosis. Indeed, blood levels of circulating MMPs may be useful for the assessment of the inflammatory activity in atherosclerosis and the prediction of cardiovascular risk. The availability of a wide variety of low-molecular MMP inhibitors that can be conjugated with various labels provides a good perspective for specific targeting of MMPs and implementation of imaging techniques to visualize MMP activity in atherosclerotic plaques and, most interestingly, to monitor responses to antiatheroslerosis therapies. Finally, because of the crucial role of ECM in cardiovascular repair, the regenerative potential of ECM could be successfully used in constructing engineered scaffolds and vessels that mimic properties of the natural ECM and consist of the native ECM components or composite biomaterials. These scaffolds possess a great promise in vascular tissue engineering.
Collapse
|
18
|
Wang Y, Lei R, Zhuang X, Zhang N, Pan H, Li G, Hu J, Pan X, Tao Q, Fu D, Xiao J, Chin YE, Kang Y, Yang Q, Hu G. DLC1-dependent parathyroid hormone-like hormone inhibition suppresses breast cancer bone metastasis. J Clin Invest 2014; 124:1646-59. [PMID: 24590291 DOI: 10.1172/jci71812] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 12/20/2013] [Indexed: 12/27/2022] Open
Abstract
Bone metastasis is a frequent complication of breast cancer that is often accelerated by TGF-β signaling; however, little is known about how the TGF-β pathway is regulated during bone metastasis. Here we report that deleted in liver cancer 1 (DLC1) is an important regulator of TGF-β responses and osteolytic metastasis of breast cancer cells. In murine models, breast cancer cells lacking DLC1 expression exhibited enhanced capabilities of bone metastasis. Knockdown of DLC1 in cancer cells promoted bone metastasis, leading to manifested osteolysis and accelerated death in mice, while DLC1 overexpression suppressed bone metastasis. Activation of Rho-ROCK signaling in the absence of DLC1 mediated SMAD3 linker region phosphorylation and TGF-β-induced expression of parathyroid hormone-like hormone (PTHLH), leading to osteoclast maturation for osteolytic colonization. Furthermore, pharmacological inhibition of Rho-ROCK effectively reduced PTHLH production and breast cancer bone metastasis in vitro and in vivo. Evaluation of clinical breast tumor samples revealed that reduced DLC1 expression was linked to elevated PTHLH expression and organ-specific metastasis to bone. Overall, our findings define a stroma-dependent paradigm of Rho signaling in cancer and implicate Rho-TGF-β crosstalk in osteolytic bone metastasis.
Collapse
|
19
|
|
20
|
Kleinmann N, Duivenvoorden WCM, Hopmans SN, Beatty LK, Qiao S, Gallino D, Lhotak S, Daya D, Paschos A, Austin RC, Pinthus JH. Underactivation of the adiponectin-adiponectin receptor 1 axis in clear cell renal cell carcinoma: implications for progression. Clin Exp Metastasis 2013; 31:169-83. [PMID: 24096711 DOI: 10.1007/s10585-013-9618-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 09/17/2013] [Indexed: 12/11/2022]
Abstract
Energy-sensing pathways, normally coordinated by 5' AMP-activated protein kinase (AMPK), are dysregulated in renal cell carcinoma (RCC). Obesity can accentuate the pre-existing pro-tumorigenic metabolic machinery in RCC cells through its associated obesogenic hormonal milieu, characterized by lower circulating levels of adiponectin. In RCC patients, low adiponectin levels associate clinically with more aggressive disease. We investigated the adiponectin signaling pathway in RCC, focusing on adiponectin receptor 1 (AdipoR1) and associated activation of AMPK. AdipoR1 protein in RCC and normal surrounding renal tissues was determined by Western blot analysis and immunohistochemistry. Anti-tumorigenic effects of adiponectin in RCC cells in vitro were investigated via VEGF and MMP ELISA and invasion assays. Using in vivo models of RCC, the effect of AdipoR1-knockdown (shRNA) on tumor latency, growth and dissemination were determined. AdipoR1 protein was significantly reduced in clear cell RCC specimens. Adiponectin treatment inhibited VEGF, MMP-2 and MMP-9 secretion and activity and invasive and migratory capacities of RCC cells. AMPKα1-knockdown (shRNA) attenuated adiponectin's effects. In cells stably expressing AdipoR1-specific shRNA, AMPK activation by adiponectin was significantly reduced compared to cells expressing control shRNA. In vivo, AdipoR1 knockdown increased the growth, dissemination and angiogenesis of RCC. These findings suggest that deficiencies in the entire adiponectin hormonal axis (the hormone and its receptor) result in underactivation of AMPK leading to increased angiogenic and invasive capacities of RCC. The established link between obesity and RCC can therefore be further explained by the adiponectin deficiency in obese individuals together with reduced AdipoR1 protein in RCC.
Collapse
Affiliation(s)
- Nir Kleinmann
- Division of Urology, Department of Surgery, McMaster University, Hamilton, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Msaouel P, Nandikolla G, Pneumaticos SG, Koutsilieris M. Bone microenvironment-targeted manipulations for the treatment of osteoblastic metastasis in castration-resistant prostate cancer. Expert Opin Investig Drugs 2013; 22:1385-400. [PMID: 24024652 DOI: 10.1517/13543784.2013.824422] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Most patients with advanced prostate cancer will develop incurable bone metastasis. Although prostate cancer is the quintessential androgen-dependent neoplastic disease in males, the tumor will ultimately become refractory to androgen ablation treatment. Understanding the complex dialog between prostate cancer and the bone microenvironment has allowed the development of promising treatment strategies. AREAS COVERED The present review summarizes the pathophysiology of prostate cancer bone metastasis and provides a concise update on bone microenvironment-targeted therapies for prostate cancer. The current and future prospects and challenges of these strategies are also discussed. EXPERT OPINION A wide variety of signaling pathways, bone turnover homeostatic mechanisms and immunoregulatory networks are potential targets for the treatment of metastatic castration-resistant prostate cancer (mCRPC). Anti-survival factor therapy can enhance the efficacy of existing treatment regimens for mCRPC by exploiting the interaction between the bone microenvironment and androgen signaling networks. In addition, many novel bone microenvironment-targeted strategies have produced promising objective clinical responses. Further elucidation of the complex interactions between prostate cancer cells and the bone stroma will open up new avenues for treatment interventions that can produce sustained cancer suppression.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Jacobi Medical Center, Department of Internal Medicine, Albert Einstein College of Medicine , Bronx, NY , USA
| | | | | | | |
Collapse
|
22
|
Sengupta S, Jana S, Biswas S, Mandal PK, Bhattacharyya A. Cooperative involvement of NFAT and SnoN mediates transforming growth factor-β (TGF-β) induced EMT in metastatic breast cancer (MDA-MB 231) cells. Clin Exp Metastasis 2013; 30:1019-31. [PMID: 23832742 DOI: 10.1007/s10585-013-9600-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 06/27/2013] [Indexed: 12/17/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a secondary phenomenon concomitantly associated with the tumor progression. The regulatory signals and mechanistic details of EMT are not fully elucidated. Here, we shared a TGF-β mediated mechanism of EMT in breast cancer (MDA-MB 231) cells. Initial exposure of TGF-β for 48 h, enhanced the rate of cell proliferation and associated with EMT of MDA-MB 231 cells. The EMT was characterized by observing the increased N-cadherin, fibronectin, Snail expression and associated with the morphological change with a reduced E-cadherin expression. NFAT, a transcription factor, alters tumor suppressive function of TGF-β towards tumor progression. Up regulation of NFAT, coupled with a foremost translocation of one oncogenic protein SnoN from cytoplasm to nucleus was noticed during this TGF-β mediated EMT. Silencing of NFAT also showed the inhibition of TGF-β mediated EMT characterized by down regulation of N-cadherin and associated with reduced expression of SnoN. In addition, it was also observed that NFAT sequestering the Smad3 prevents the proteasome mediated degradation of SnoN and this SnoN has a role on the regulation of MMP-2, MMP-9 activity. Increased Smad3-SnoN interaction and proteasome mediated degradation of SnoN were detected after silencing of NFAT with a reduced MMP-2, MMP-9 activity. All of these observations provide a fresh mechanism in which by a twofold involvement of NFAT and SnoN plays a crucial role in TGF-β mediated EMT by recruiting the effector molecules N-cadherin and MMP-2, MMP-9.
Collapse
Affiliation(s)
- Suman Sengupta
- Immunology Lab, Department of Zoology, University of Calcutta, Kolkata, 700019, West Bengal, India
| | | | | | | | | |
Collapse
|
23
|
LU QIANG, LV GANG, KIM ANDRE, HA JONGMYUNG, KIM SUHKMAN. Expression and clinical significance of extracellular matrix metalloproteinase inducer, EMMPRIN/CD147, in human osteosarcoma. Oncol Lett 2013; 5:201-207. [PMID: 23255920 PMCID: PMC3525466 DOI: 10.3892/ol.2012.981] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/26/2012] [Indexed: 01/15/2023] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor in children and adolescents. Recent studies have shown that extracellular matrix metalloproteinase inducer (EMMPRIN/CD147) promotes adhesion, invasion and metastasis of malignant tumor cells. The aim of this study was to investigate the impact of EMMPRIN/CD147 expression on prognosis and its correlation with clinicopathological characteristics in patients with osteosarcoma. The expression of EMMPRIN/CD147 in 55 surgical specimens from patients with osteosarcoma at stage IIA or above, 15 non-tumor rib bone tissues, three human osteosarcoma cell lines (Saos-2, U-2OS and MG-63), the human osteoblast cell line HOB and the malignant melanoma cell line A375 were examined by immunohistochemistry, western blot analysis and ELISA, respectively. The potential association of the levels of EMMPRIN/CD147 expression in osteosarcoma specimens with the overall survival of patients was statistically analyzed. We found that the EMMPRIN/CD147 was expressed in 45 out of 55 osteosarcomas, with immunoreactivity primarily within the membrane and cytoplasm of tumor cells, but not in the non-tumor bone tissues. We also observed that EMMPRIN/CD147 was expressed in Saos-2, U-2OS, MG-63 and A375, but not in HOB cells. The levels of EMMPRIN/CD147 expression correlated positively with the pathological degree of osteosarcoma and negatively with the survival period of patients with osteosarcoma. The expression of EMMPRIN/CD147 is a potential factor in the development and prognosis of osteosarcoma and may be a novel therapeutic target of human osteosarcoma.
Collapse
Affiliation(s)
- QIANG LU
- Department of Orthopaedics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001,
P.R. China
| | - GANG LV
- Department of Orthopaedics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001,
P.R. China
- Correspondence to: Professor Gang Lv, Department of Orthopaedics, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping, Shenyang, Liaoning 110001, P.R. China, E-mail:
| | - ANDRE KIM
- Department of Pharmaceutical Engineering, College of Medical Life Sciences, Silla University, Busan 617-736
| | - JONG-MYUNG HA
- Department of Pharmaceutical Engineering, College of Medical Life Sciences, Silla University, Busan 617-736
| | - SUHKMAN KIM
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 609-735,
Republic of Korea
| |
Collapse
|
24
|
Bone metastasis in breast cancer: the story of RANK-ligand. J Egypt Natl Canc Inst 2012; 24:107-14. [PMID: 22929916 DOI: 10.1016/j.jnci.2012.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 06/05/2012] [Accepted: 06/10/2012] [Indexed: 11/22/2022] Open
Abstract
The primary cellular mechanism responsible for osteolytic bone metastases is osteoclastic activation. Preclinical models have shown that breast cancer cells can produce parathyroid hormone-related protein (PTHrP), and other osteolytic molecules, which stimulate excessive osteoclastic bone resorption and establishment of osteolytic lesions. It has been shown that PTHrP by itself cannot directly induce osteoclastic activation, but it mediates its effect through the transactivation of RANK-ligand (RANKL) gene on stromal and osteoblastic cells. Accordingly RANKL up-regulation has been considered as a prerequisite in virtually all conditions of cancer induced bone destruction. Hence, therapeutic targeting of RANKL seems to be a rational approach to treat or even to prevent the process of bone metastases. In this review, we will focus on the unique patho-physiological aspects related to the evolution of bone metastases in breast cancer, emphasizing the pivotal role of RANKL and some other key molecules in osteoclastic bone resorption. We will discuss the therapeutic interventions using bisphosphonates and RANKL inhibitors in patients with bone metastases and the outcome of this novel approach.
Collapse
|
25
|
Abstract
Background Accelerated bone loss in patients with cancer is a frequent problem that may result from invasion of the cancer to bone, paraneoplastic tumor proteins, and/or hormonal therapies utilized for cancer treatment. Patients with osteolytic bone disease from multiple myeloma and bone metastases from solid tumors may develop a vicious cycle of bone destruction involving both osteolytic and osteoblastic effects. Consequently, a variety of skeletal-related events (SREs) may occur, including pathological fractures, hypercalcemia, spinal cord compression, and the need for surgical intervention and radiation therapy. Methods This article reviews the results of trials that investigated the safety and efficacy of pharmacologic agents, including bisphosphonates and denosumab, for treatment of bone metastases. This analysis is derived from an assessment of the medical literature. Results Beneficial systemic therapies for bone metastases have been developed to decrease SREs. Possible antitumor effects of the bisphosphonates are explored. In addition, the utility of markers of bone turnover in relation to response to therapy and survival, the safety and toxicity of bone-targeted therapies, treatment guidelines, and economic considerations are also discussed. Conclusions Effective systemic therapies for metastatic bone disease are available. Ongoing and future research projects in this field are also presented.
Collapse
Affiliation(s)
- Loretta S. Loftus
- Comprehensive Breast Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Sophia Edwards-Bennett
- Department of Radiation Oncology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Gerald H. Sokol
- Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
26
|
TGF-β1 modulates the homeostasis between MMPs and MMP inhibitors through p38 MAPK and ERK1/2 in highly invasive breast cancer cells. BMC Cancer 2012; 12:26. [PMID: 22260435 PMCID: PMC3277461 DOI: 10.1186/1471-2407-12-26] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/19/2012] [Indexed: 11/21/2022] Open
Abstract
Background Metastasis is the main factor responsible for death in breast cancer patients. Matrix metalloproteinases (MMPs) and their inhibitors, known as tissue inhibitors of MMPs (TIMPs), and the membrane-associated MMP inhibitor (RECK), are essential for the metastatic process. We have previously shown a positive correlation between MMPs and their inhibitors expression during breast cancer progression; however, the molecular mechanisms underlying this coordinate regulation remain unknown. In this report, we investigated whether TGF-β1 could be a common regulator for MMPs, TIMPs and RECK in human breast cancer cell models. Methods The mRNA expression levels of TGF-β isoforms and their receptors were analyzed by qRT-PCR in a panel of five human breast cancer cell lines displaying different degrees of invasiveness and metastatic potential. The highly invasive MDA-MB-231 cell line was treated with different concentrations of recombinant TGF-β1 and also with pharmacological inhibitors of p38 MAPK and ERK1/2. The migratory and invasive potential of these treated cells were examined in vitro by transwell assays. Results In general, TGF-β2, TβRI and TβRII are over-expressed in more aggressive cells, except for TβRI, which was also highly expressed in ZR-75-1 cells. In addition, TGF-β1-treated MDA-MB-231 cells presented significantly increased mRNA expression of MMP-2, MMP-9, MMP-14, TIMP-2 and RECK. TGF-β1 also increased TIMP-2, MMP-2 and MMP-9 protein levels but downregulated RECK expression. Furthermore, we analyzed the involvement of p38 MAPK and ERK1/2, representing two well established Smad-independent pathways, in the proposed mechanism. Inhibition of p38MAPK blocked TGF-β1-increased mRNA expression of all MMPs and MMP inhibitors analyzed, and prevented TGF-β1 upregulation of TIMP-2 and MMP-2 proteins. Moreover, ERK1/2 inhibition increased RECK and prevented the TGF-β1 induction of pro-MMP-9 and TIMP-2 proteins. TGF-β1-enhanced migration and invasion capacities were blocked by p38MAPK, ERK1/2 and MMP inhibitors. Conclusion Altogether, our results support that TGF-β1 modulates the mRNA and protein levels of MMPs (MMP-2 and MMP-9) as much as their inhibitors (TIMP-2 and RECK). Therefore, this cytokine plays a crucial role in breast cancer progression by modulating key elements of ECM homeostasis control. Thus, although the complexity of this signaling network, TGF-β1 still remains a promising target for breast cancer treatment.
Collapse
|
27
|
Zeng W, Wan R, Zheng Y, Singh SR, Wei Y. Hypoxia, stem cells and bone tumor. Cancer Lett 2011; 313:129-36. [PMID: 21999934 PMCID: PMC3215823 DOI: 10.1016/j.canlet.2011.09.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 09/20/2011] [Accepted: 09/21/2011] [Indexed: 12/26/2022]
Abstract
Normal oxygen level is critical for niches that together with other components of the niche play vital role in regulating stem or tumor cells behavior. Hypoxia plays an important role in normal development and disease progression, including the growth of solid tumors. The hypoxia inducible factors (HIFs) are the key mediators of the cellular response to hypoxia. In this review, we focused on the role of HIFs on bone tumor formation. Further, we also emphasized how hypoxia, stem cells, and its niches regulate the bone tumorigenesis.
Collapse
Affiliation(s)
- Wen Zeng
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Shanghai First People’s Hospital, Shanghai Jiaotong University, 200080, People’s Republic of China
| | - Rong Wan
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Yuehuan Zheng
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Shree Ram Singh
- Mouse Cancer Genetics Program, National Institutes of Health, National Cancer Institute at Frederick, Frederick, Maryland, 21702, USA
| | - Yiyong Wei
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| |
Collapse
|
28
|
Sottnik JL, Zhang J, Macoska JA, Keller ET. The PCa Tumor Microenvironment. CANCER MICROENVIRONMENT 2011; 4:283-97. [PMID: 21728070 DOI: 10.1007/s12307-011-0073-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 06/23/2011] [Indexed: 12/31/2022]
Abstract
The tumor microenvironment (TME) is a very complex niche that consists of multiple cell types, supportive matrix and soluble factors. Cells in the TME consist of both host cells that are present at tumor site at the onset of tumor growth and cells that are recruited in either response to tumor- or host-derived factors. PCa (PCa) thrives on crosstalk between tumor cells and the TME. Crosstalk results in an orchestrated evolution of both the tumor and microenvironment as the tumor progresses. The TME reacts to PCa-produced soluble factors as well as direct interaction with PCa cells. In return, the TME produces soluble factors, structural support and direct contact interactions that influence the establishment and progression of PCa. In this review, we focus on the host side of the equation to provide a foundation for understanding how different aspects of the TME contribute to PCa progression. We discuss immune effector cells, specialized niches, such as the vascular and bone marrow, and several key protein factors that mediate host effects on PCa. This discussion highlights the concept that the TME offers a potentially very fertile target for PCa therapy.
Collapse
Affiliation(s)
- Joseph L Sottnik
- Department of Urology, University of Michigan, RM 5308 CC, Ann Arbor, MI, 48109-8940, USA
| | | | | | | |
Collapse
|
29
|
Wendt MK, Tian M, Schiemann WP. Deconstructing the mechanisms and consequences of TGF-β-induced EMT during cancer progression. Cell Tissue Res 2011; 347:85-101. [PMID: 21691718 DOI: 10.1007/s00441-011-1199-1] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 06/01/2011] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-β (TGF-β) is a potent pleiotropic cytokine that regulates mammalian development, differentiation, and homeostasis in essentially all cell types and tissues. TGF-β normally exerts anticancer activities by prohibiting cell proliferation and by creating cell microenvironments that inhibit cell motility, invasion, and metastasis. However, accumulating evidence indicates that the process of tumorigenesis, particularly that associated with metastatic progression, confers TGF-β with oncogenic activities, a functional switch known as the "TGF-β paradox." The molecular determinants governing the TGF-β paradox are complex and represent an intense area of investigation by researchers in academic and industrial settings. Recent findings link genetic and epigenetic events in mediating the acquisition of oncogenic activity by TGF-β, as do aberrant alterations within tumor microenvironments. These events coalesce to enable TGF-β to direct metastatic progression via the stimulation of epithelial-mesenchymal transition (EMT), which permits carcinoma cells to abandon polarized epithelial phenotypes in favor of apolar mesenchymal-like phenotypes. Attempts to deconstruct the EMT process induced by TGF-β have identified numerous signaling molecules, transcription factors, and microRNAs operant in mediating the initiation and resolution of this complex transdifferentiation event. In addition to its ability to enhance carcinoma cell invasion and metastasis, EMT also endows transitioned cells with stem-like properties, including the acquisition of self-renewal and tumor-initiating capabilities coupled to chemoresistance. Here, we review recent findings that delineate the pathophysiological mechanisms whereby EMT stimulated by TGF-β promotes metastatic progression and disease recurrence in human carcinomas.
Collapse
Affiliation(s)
- Michael K Wendt
- Case Comprehensive Cancer Center, Division of General Medical Sciences-Oncology, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
30
|
Onishi T, Hayashi N, Theriault RL, Hortobagyi GN, Ueno NT. Future directions of bone-targeted therapy for metastatic breast cancer. Nat Rev Clin Oncol 2010; 7:641-51. [PMID: 20808302 DOI: 10.1038/nrclinonc.2010.134] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bone is the most common metastatic site for breast cancer, and bone metastases can cause pain as well as risk of pathological fractures. Emerging treatments for metastatic bone disease have arisen from advances in our understanding of the unique cellular and molecular mechanisms that contribute to bone metastasis. The interaction between tumor cells and the bone microenvironment results in a 'vicious cycle' that increases both bone destruction and tumor burden. The tumor secretes factors, such as parathyroid hormone-related peptide, that stimulate osteoclastogenesis. Similarly, the bone stroma produces growth factors, such as transforming growth factor β, that promote tumor growth in bone. Therapeutic targeting of these microenvironmental factors is under intensive investigation. Other attractive therapeutic targets include signaling molecules, such as receptor activator of nuclear factor κB ligand, Src kinase, and cathepsin K, all of which regulate osteoclast function, and chemokine receptor 4, which is involved in the homing of tumor cells to bone. In this Review, we describe the progress and future directions of novel bone-targeted therapies that may reduce or prevent destructive bone metastasis from breast cancer. Novel modalities for predicting and monitoring treatment response will also be described.
Collapse
Affiliation(s)
- Tomifumi Onishi
- Christus Hospital-St Elizabeth, 2830 Calder Street, Beaumont, TX 77702, USA
| | | | | | | | | |
Collapse
|
31
|
Taylor MA, Parvani JG, Schiemann WP. The pathophysiology of epithelial-mesenchymal transition induced by transforming growth factor-beta in normal and malignant mammary epithelial cells. J Mammary Gland Biol Neoplasia 2010; 15:169-90. [PMID: 20467795 PMCID: PMC3721368 DOI: 10.1007/s10911-010-9181-1] [Citation(s) in RCA: 183] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Accepted: 04/22/2010] [Indexed: 12/14/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is an essential process that drives polarized, immotile mammary epithelial cells (MECs) to acquire apolar, highly migratory fibroblastoid-like features. EMT is an indispensable process that is associated with normal tissue development and organogenesis, as well as with tissue remodeling and wound healing. In stark contrast, inappropriate reactivation of EMT readily contributes to the development of a variety of human pathologies, particularly those associated with tissue fibrosis and cancer cell invasion and metastasis, including that by breast cancer cells. Although metastasis is unequivocally the most lethal aspect of breast cancer and the most prominent feature associated with disease recurrence, the molecular mechanisms whereby EMT mediates the initiation and resolution of breast cancer metastasis remains poorly understood. Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine that is intimately involved in regulating numerous physiological processes, including cellular differentiation, homeostasis, and EMT. In addition, TGF-beta also functions as a powerful tumor suppressor in MECs, whose neoplastic development ultimately converts TGF-beta into an oncogenic cytokine in aggressive late-stage mammary tumors. Recent findings have implicated the process of EMT in mediating the functional conversion of TGF-beta during breast cancer progression, suggesting that the chemotherapeutic targeting of EMT induced by TGF-beta may offer new inroads in ameliorating metastatic disease in breast cancer patients. Here we review the molecular, cellular, and microenvironmental factors that contribute to the pathophysiological activities of TGF-beta during its regulation of EMT in normal and malignant MECs.
Collapse
Affiliation(s)
- Molly A Taylor
- Case Comprehensive Cancer Center, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
32
|
Wendt MK, Allington TM, Schiemann WP. Mechanisms of the epithelial-mesenchymal transition by TGF-beta. Future Oncol 2010; 5:1145-68. [PMID: 19852727 DOI: 10.2217/fon.09.90] [Citation(s) in RCA: 252] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The formation of epithelial cell barriers results from the defined spatiotemporal differentiation of stem cells into a specialized and polarized epithelium, a process termed mesenchymal-epithelial transition. The reverse process, epithelial-mesenchymal transition (EMT), is a metastable process that enables polarized epithelial cells to acquire a motile fibroblastoid phenotype. Physiological EMT also plays an essential role in promoting tissue healing, remodeling or repair in response to a variety of pathological insults. On the other hand, pathophysiological EMT is a critical step in mediating the acquisition of metastatic phenotypes by localized carcinomas. Although metastasis clearly is the most lethal aspect of cancer, our knowledge of the molecular events that govern its development, including those underlying EMT, remain relatively undefined. Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine that oversees and directs all aspects of cell development, differentiation and homeostasis, as well as suppresses their uncontrolled proliferation and transformation. Quite dichotomously, tumorigenesis subverts the tumor suppressing function of TGF-beta, and in doing so, converts TGF-beta to a tumor promoter that stimulates pathophysiological EMT and metastasis. It therefore stands to reason that determining how TGF-beta induces EMT in developing neoplasms will enable science and medicine to produce novel pharmacological agents capable of preventing its ability to do so, thereby improving the clinical course of cancer patients. Here we review the cellular, molecular and microenvironmental mechanisms used by TGF-beta to mediate its stimulation of EMT in normal and malignant cells.
Collapse
Affiliation(s)
- Michael K Wendt
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | | | | |
Collapse
|
33
|
Hypoxia and TGF-beta drive breast cancer bone metastases through parallel signaling pathways in tumor cells and the bone microenvironment. PLoS One 2009; 4:e6896. [PMID: 19727403 PMCID: PMC2731927 DOI: 10.1371/journal.pone.0006896] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2009] [Accepted: 08/16/2009] [Indexed: 12/27/2022] Open
Abstract
Background Most patients with advanced breast cancer develop bone metastases, which cause pain, hypercalcemia, fractures, nerve compression and paralysis. Chemotherapy causes further bone loss, and bone-specific treatments are only palliative. Multiple tumor-secreted factors act on the bone microenvironment to drive a feed-forward cycle of tumor growth. Effective treatment requires inhibiting upstream regulators of groups of prometastatic factors. Two central regulators are hypoxia and transforming growth factor (TGF)- β. We asked whether hypoxia (via HIF-1α) and TGF-β signaling promote bone metastases independently or synergistically, and we tested molecular versus pharmacological inhibition strategies in an animal model. Methodology/Principal Findings We analyzed interactions between HIF-1α and TGF-β pathways in MDA-MB-231 breast cancer cells. Only vascular endothelial growth factor (VEGF) and the CXC chemokine receptor 4 (CXCR4), of 16 genes tested, were additively increased by both TGF-β and hypoxia, with effects on the proximal promoters. We inhibited HIF-1α and TGF-β pathways in tumor cells by shRNA and dominant negative receptor approaches. Inhibition of either pathway decreased bone metastasis, with no further effect of double blockade. We tested pharmacologic inhibitors of the pathways, which target both the tumor and the bone microenvironment. Unlike molecular blockade, combined drug treatment decreased bone metastases more than either alone, with effects on bone to decrease osteoclastic bone resorption and increase osteoblast activity, in addition to actions on tumor cells. Conclusions/Significance Hypoxia and TGF-β signaling in parallel drive tumor bone metastases and regulate a common set of tumor genes. In contrast, small molecule inhibitors, by acting on both tumor cells and the bone microenvironment, additively decrease tumor burden, while improving skeletal quality. Our studies suggest that inhibitors of HIF-1α and TGF-β may improve treatment of bone metastases and increase survival.
Collapse
|
34
|
Justulin Jr LA, Acquaro C, Carvalho RF, Silva MDP, Felisbino SL. Combined effect of the finasteride and doxazosin on rat ventral prostate morphology and physiology. ACTA ACUST UNITED AC 2009; 33:489-99. [DOI: 10.1111/j.1365-2605.2009.00963.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
35
|
Ichiyama T, Kajimoto M, Hasegawa M, Hashimoto K, Matsubara T, Furukawa S. Cysteinyl leukotrienes enhance tumour necrosis factor-alpha-induced matrix metalloproteinase-9 in human monocytes/macrophages. Clin Exp Allergy 2007; 37:608-14. [PMID: 17430359 DOI: 10.1111/j.1365-2222.2007.02692.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Matrix metalloproteinase-9 (MMP-9) is an important enzyme responsible for airway remodelling. Monocytes/macrophages have a cysteinyl leukotriene 1 (cysLT1) receptor, but its function is poorly understood. OBJECTIVE To elucidate the function of the cysLT1 receptor of human monocytes/macrophages in MMP-9 production. METHODS We examined the effect of cysLTs (LTC4, -D4 and -E4) on TNF-alpha-induced MMP-9 production in THP-1 cells, a human monocytic leukaemia cell line and peripheral blood CD14+ monocytes/macrophages. In addition, we examined the effect of pranlukast, a cysLT1 receptor antagonist, on the enhancement of TNF-alpha-induced MMP-9 production by cysLTs. RESULTS ELISA revealed that LTC4 and -D4, but not -E4, enhanced TNF-alpha-induced MMP-9 production in THP-1 cells and peripheral blood CD14+ monocytes/macrophages. Real-time polymerase chain reaction demonstrated that LTC4 and -D4, but not -E4, increased MMP-9 mRNA expression induced by TNF-alpha in THP-1 cells. Moreover, we demonstrated that pranlukast completely inhibited the enhancement of TNF-alpha-induced MMP-9 production by LTC4 and -D4 in THP-1 cells and peripheral blood CD14+ monocytes/macrophages. CONCLUSION LTC4 and -D4 enhanced the TNF-alpha-induced MMP-9 production via binding the cysLT1 receptor in human monocytes/macrophages. Pranlukast inhibited the enhancements by LTC4 and D4.
Collapse
Affiliation(s)
- T Ichiyama
- Department of Pediatrics, Yamaguchi University School of Medicine, Yamaguchi, Japan.
| | | | | | | | | | | |
Collapse
|
36
|
Choi YD, Cho NH, Ahn HS, Cho KS, Cho SY, Yang WJ. Matrix Metalloproteinase Expression in the Recurrence of Superficial Low Grade Bladder Transitional Cell Carcinoma. J Urol 2007; 177:1174-8. [PMID: 17296440 DOI: 10.1016/j.juro.2006.10.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Indexed: 11/26/2022]
Abstract
PURPOSE Superficial bladder transitional cell carcinoma has a high recurrence rate following endoscopic resection. Most parameters used to predict recurrence are associated with high grade transitional cell carcinoma. However, there are few valid parameters for predicting low grade transitional cell carcinoma recurrence. We elucidated the prime factor in recurrent superficial low grade bladder transitional cell carcinoma. MATERIALS AND METHODS A total of 40 cases of superficial low grade bladder transitional cell carcinoma were analyzed, including 20 recurrent and 20 nonrecurrent cases. Fresh frozen tumor and normal tissues were used. Real-time reverse transcriptase-polymerase chain reaction and Western blots were performed subsequently to evaluate the validity of molecules identified by 0.12K cDNA array composed of matrix metalloproteinase, oncogenes and cell cycle related genes. RESULTS On cDNA microarray analysis matrix metalloproteinase-1, 2, 9, 12 and 15, transforming growth factor-beta1, vascular endothelial growth factor and fos were found to be significantly up-regulated in recurrent cases. On real-time reverse transcriptase-polymerase chain reaction to validate those molecules matrix metalloproteinase-1 and 12, transforming growth factor-beta1, vascular endothelial growth factor and fos were significantly over expressed in recurrent cases. Western blot showed significant over expression of matrix metalloproteinase-1 and 12, transforming growth factor-beta1, vascular endothelial growth factor and fos in recurrent cases. CONCLUSIONS Over expression of matrix metalloproteinase-1 and 12, transforming growth factor-beta1, vascular endothelial growth factor and fos may be associated with the recurrence of superficial low grade bladder transitional cell carcinoma.
Collapse
Affiliation(s)
- Young Deuk Choi
- Department of Urology and Urological Science Institute, Seoul, Korea.
| | | | | | | | | | | |
Collapse
|
37
|
Celotti F, Colciago A, Negri-Cesi P, Pravettoni A, Zaninetti R, Sacchi MC. Effect of platelet-rich plasma on migration and proliferation of SaOS-2 osteoblasts: role of platelet-derived growth factor and transforming growth factor-beta. Wound Repair Regen 2006; 14:195-202. [PMID: 16630109 DOI: 10.1111/j.1743-6109.2006.00110.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Platelet-enriched plasma (PRP) is used in therapy as a source of growth factors in bone fracture and wound healing; however, few data exist on its role in the different aspects of the healing process. The effect of PRP and of the two main growth factors present in this preparation (platelet-derived growth factor [PDGF] and transforming growth factor-beta [TGF-beta]) was evaluated in vitro using the human osteoblastic cell line SaOS-2, which was shown by reverse transcription-polymerase chain reaction to express both PDGF-alpha and -beta receptors. Batroxobine-activated PRP was added in different concentrations to SaOS-2 cells to assess cell migration (by a microchemotaxis assay) and cell proliferation (by [3H]-thymidine incorporation into the DNA). Immunoneutralization with anti-PDGF-beta or anti-TGF-beta antibodies allowed the assessment of the specific role of these growth factors. The overall results obtained indicate that PRP dose-dependently stimulates both chemotaxis and cell proliferation. PDGF and TGF-beta appear to exert distinct effects on the two parameters, the former involved in stimulating cell migration and the latter in inhibiting cell proliferation. It is concluded that the different growth factors present in activated PRP can specifically contribute to the main processes of tissue regeneration.
Collapse
Affiliation(s)
- Fabio Celotti
- Institute of Endocrinology, University of Milano, Milano, Italy.
| | | | | | | | | | | |
Collapse
|
38
|
Stark AM, Anuszkiewicz B, Mentlein R, Yoneda T, Mehdorn HM, Held-Feindt J. Differential expression of matrix metalloproteinases in brain- and bone-seeking clones of metastatic MDA-MB-231 breast cancer cells. J Neurooncol 2006; 81:39-48. [PMID: 16850107 DOI: 10.1007/s11060-006-9207-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Accepted: 05/31/2006] [Indexed: 10/24/2022]
Abstract
Matrix Metalloproteinases (MMPs) play a crucial role in breast cancer metastasis. We examined the mRNA and protein expression of several MMPs in brain- and bone-seeking clones of MDA-MB-231 breast cancer cells, their transcriptional regulation and their functional role in the metastatic process. MMP mRNA expression was examined using real-time reverse transcription polymerase chain reaction. Protein expression was examined using enzyme linked immunosorbent essay (ELISA). The inducibility of mRNA and protein expression was tested with TPA (phorbol 12-myristate 13-acetate; 50 microM); epidermal growth factor and transforming growth factor beta (20 ng/ml both). Migration and invasion assays were performed with the QCM 96-Well Migration/Invasion Assay (8 microm; Chemicon) over 24 h with or without specific MMPs inhibitors (MMP Inhibitor I Mix (5 microM); MMP-2/MMP-9 Inhibitor III (50 microM); EMD Biosciences). We found significantly higher mRNA expression of MMP-1 and -9 in brain-seeking 231-clones in comparison to -bone and -parental cells. In contrast, the mRNA expression of MMP-3 and -14 was comparable in all cells lines examined and MMP-13 expression was lower in both selective metastatic lines. MMP-2 and -8 were not expressed. ELISA revealed a higher amount of total as well as active MMP-1 and -9 in brain-seeking cells. TPA stimulation showed that MMP-1 and -9 transcription was inducible on the mRNA and protein level in 231-parental but not in 231-brain or -bone. 231-brain showed the highest migration and invasive capacity which could be decreased by the application of MMP-1 and/or MMP-9 inhibitor. Our results indicate functional importance of MMP-1 and -9 overexpression in brain metastasis in an in vitro model.
Collapse
Affiliation(s)
- Andreas M Stark
- Department of Neurosurgery, University of Kiel, Schittenhelmstrasse 10, 24105 Kiel, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Liu G, Zhang F, Lee J, Dong Z. Selective induction of interleukin-8 expression in metastatic melanoma cells by transforming growth factor-beta 1. Cytokine 2006; 31:241-9. [PMID: 15975819 DOI: 10.1016/j.cyto.2005.03.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Revised: 02/11/2005] [Accepted: 03/11/2005] [Indexed: 11/22/2022]
Abstract
Interleukin (IL)-8 and transforming growth factor (TGF)-beta1 are proangiogenic factors overexpressed in advanced human melanoma. We investigated the effects of TGF-beta1 on IL-8 expression in the well-characterized A375 human melanoma system. We demonstrated by enzyme-linked immunoassay and Northern blot analysis that TGF-beta1 selectively induced IL-8 expression, at both protein and mRNA levels, in highly metastatic A375SM cells but not cells of their poorly metastatic parental line A375P. Transient transfection with luciferase reporter gene constructs revealed that TGF-beta1 activated IL-8 promoter activity in A375SM cells but not A375P cells. Studies with progressive 5' deletion constructs and site-specific mutations demonstrated that a construct containing -133 to +44 of the 5'-flanking sequence was necessary and sufficient for maximal TGF-beta1-induced transcription response and that TGF-beta1-induced activation of IL-8 promoter depended on AP-1 (-126 to -120 bp), NF-kappaB (-94 to -71 bp), and C/EBP-like factor NF-IL6 (-94 to -81 bp) in this region. Interestingly, both A375P and A375SM cells expressed type I and type II TGF-beta receptors and TGF-beta1 induced the nuclear translocation of Smad3 protein in both A375P and A375SM cells. Moreover, both A375P and A375SM cells were susceptible to TGF-beta1-induced growth inhibition. Our data thus demonstrated that TGF-beta1 selectively induced IL-8 expression in highly metastatic A375SM melanoma cells. This TGF-beta1-induced IL-8 expression could be an amplification cascade responsible for overexpression of IL-8 in human melanoma and one of potential mechanisms by which TGF-beta1 promotes angiogenesis, growth, and metastasis of human melanoma.
Collapse
Affiliation(s)
- Guozhen Liu
- Department of Infectious Diseases, XiangYa Hospital, Central-South University, Chang Sha 410008, People's Republic of China
| | | | | | | |
Collapse
|
40
|
Kukacka J, Průsa R, Kotaska K, Pelouch V. Matrix metalloproteinases and their function in myocardium. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2005; 149:225-36. [PMID: 16601761 DOI: 10.5507/bp.2005.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A significant number of myocardial diseases are accompanied by increased synthesis and degradation of the extracellular matrix (ECM) as well as by changed maturation and incorporation of ECM components. Important groups of enzymes responsible for both normal and pathological processes in ECM remodeling are matrix metaloproteinases (MMPs). These enzymes share a relatively conserved structure with a number of identifiable modules linked to their specific functions. The most important function of MMPs is the ability to cleave various ECM components; including such rigid molecules as fibrillar collagen molecules. The amount and activity of MMPs in cardiac tissue are regulated by a range of activating and inhibiting processes. Although MMPs play multifarious roles in many myocardial diseases, here we have focused on their function in ischemic cardiac tissue, dilated cardiomyopathy and hypertrophied cardiac tissue. The inhibition of MMPs by means of synthetic inhibitors seems to be a promising strategy in cardiac disease treatment. Their effects on diseased cardiac tissue have been successfully tested in several experimental studies.
Collapse
Affiliation(s)
- Jirí Kukacka
- Department of Clinical Biochemistry and Pathobiochemistry Faculty Hospital Motol and 2nd Medical Faculty of Charles University, Prague, Czech Republic
| | | | | | | |
Collapse
|
41
|
Saed GM, Kruger M, Diamond MP. Enhanced matrix metalloproteinase expression by Tisseel in mesothelial cells, normal peritoneal fibroblasts, and adhesion fibroblasts. EUROPEAN JOURNAL OF PLASTIC SURGERY 2005. [DOI: 10.1007/s00238-005-0006-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
42
|
Annabi B, Bouzeghrane M, Currie JC, Hawkins R, Dulude H, Daigneault L, Ruiz M, Wisniewski J, Garde S, Rabbani SA, Panchal C, Wu JJ, Béliveau R. A PSP94-derived Peptide PCK3145 inhibits MMP-9 Secretion and Triggers CD44 Cell Surface Shedding: Implication in Tumor Metastasis. Clin Exp Metastasis 2005; 22:429-39. [PMID: 16283486 DOI: 10.1007/s10585-005-2669-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Accepted: 09/01/2005] [Indexed: 10/25/2022]
Abstract
PURPOSE PCK3145 is a synthetic peptide corresponding to amino acids 31-45 of prostate secretory protein 94, which can reduce experimental skeletal metastases and prostate tumor growth in vivo. Part of its biological action involves the reduction of circulating plasma matrix metalloproteinase (MMP)-9, a crucial mediator in extracellular matrix (ECM) degradation during tumor metastasis and cancer cell invasion. The antimetastatic mechanism of action of PCK3145 is however, not understood. EXPERIMENTAL DESIGN HT-1080 fibrosarcoma cells were treated with PCK3145, and cell lysates used for immunoblot analysis of small GTPase RhoA and membrane type (MT)1-MMP protein expression. Conditioned media was used to monitor soluble MMP-9 gelatinolytic activity by zymography and protein expression by immunoblotting. RT-PCR was used to assess RhoA, MT1-MMP, MMP-9, RECK, and CD44 gene expression. Flow cytometry was used to monitor cell surface expression of CD44 and of membrane-bound MMP-9. Cell adhesion was performed on different purified ECM proteins, while cell migration was specifically performed on hyaluronic acid (HA). RESULTS We found that PCK3145 inhibited HT-1080 cell adhesion onto HA, laminin-1, and type-I collagen suggesting the common implication of the cell surface receptor CD44. In fact, PCK3145 triggered the shedding of CD44 from the cell surface into the conditioned media. PCK3145 also inhibited MMP-9 secretion and binding to the cell surface. This effect was correlated to increased RhoA and MT1-MMP gene and protein expression. CONCLUSIONS Our data suggest that PCK3145 may antagonize tumor cell metastatic processes by inhibiting both MMP-9 secretion and its potential binding to its cell surface docking receptor CD44. Such mechanism may involve RhoA signaling and increase in MT1-MMP-mediated CD44 shedding. Together with its beneficial effects in clinical trials, this is the first demonstration of PCK3145 acting as a MMP secretion inhibitor.
Collapse
Affiliation(s)
- Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Département de Chimie-Biochimie, Université du Québec à Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Aharinejad S, Krenn K, Paulus P, Schäfer R, Zuckermann A, Grimm M, Abraham D. Differential role of TGF-beta1/bFGF and ET-1 in graft fibrosis in heart failure patients. Am J Transplant 2005; 5:2185-92. [PMID: 16095497 DOI: 10.1111/j.1600-6143.2005.01006.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Collagen overproduction characteristic for dilated cardiomyopathy (DCM) is coregulated by endothelin (ET)-1, transforming growth factor (TGF)-beta1, basic fibroblast growth factor (bFGF) and matrix metalloproteases (MMPs). Whether these molecules affect grafts transplanted to heart failure patients is unknown. In 67 idiopathic DCM patients, 31 patients with ischemic cardiomyopathy (ICM) and 16 controls, the myocardial bFGF, TGF-beta1, pro-collagen (PrCol) type 1 (PrCol1-alpha1, -alpha2) and MMP expressions were examined using real-time RT-PCR or Western blotting. mRNA expression was measured in grafts for 1 year. TGF-beta1/bFGF stimulation or gene silencing was used to examine their effect on collagen synthesis in cardiac tissue cultures. TGF-beta1 and PrCol1 were upregulated in DCM only, while bFGF was upregulated in both groups versus controls. TGF-beta1 downregulated MMP-1 and upregulated collagen 1, whereas bFGF upregulated MMP-13 in DCM tissue. Post-transplant PrCol1-alpha1, -alpha2 and ET-1 mRNA increased over time in grafts of DCM patients only, while other factors returned to control baseline levels in DCM and ICM. These data indicate that cardiac transplantation corrects the dysregulated TGF/bFGF/MMP-1/MMP-13, but not the excess collagen and ET-1 synthesis in cardiac grafts transplanted to DCM patients. ET-1 might be a major pathologic trigger for graft fibrosis in DCM.
Collapse
Affiliation(s)
- Seyedhossein Aharinejad
- Department of Cardio-Thoracic Surgery, Vienna Medical University, Waehringer Guertel 18-20, A-1090 Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
44
|
Ferretti G, Fabi A, Carlini P, Papaldo P, Cordiali Fei P, Di Cosimo S, Salesi N, Giannarelli D, Alimonti A, Di Cocco B, D'Agosto G, Bordignon V, Trento E, Cognetti F. Zoledronic-acid-induced circulating level modifications of angiogenic factors, metalloproteinases and proinflammatory cytokines in metastatic breast cancer patients. Oncology 2005; 69:35-43. [PMID: 16088233 DOI: 10.1159/000087286] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2004] [Accepted: 01/10/2005] [Indexed: 12/29/2022]
Abstract
BACKGROUND To evaluate the modifications of circulating angiogenic factors, metalloproteinases and acute-phase cytokines after the first single zoledronic acid (ZA) intravenous infusion. EXPERIMENTAL DESIGN Eighteen consecutive breast cancer patients with bone metastases were evaluated for circulating levels of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), metalloproteinase 1 (MMP-1), metalloproteinase 2 (MMP-2), interleukins 1beta, 6 and 8 (IL-1beta, IL-6, IL-8), interferon gamma, tumor necrosis factor alpha (TNF-alpha) and transforming growth factor beta1 just before and 2 and 7 days after ZA infusion. RESULTS The MMP-2 basal value showed a statistically significant decrease 48 h after ZA (p = 0.01), being at 7 days higher than the day 2 value (p = 0.03). The VEGF basal value showed a statistically significant decrease 48 h after ZA infusion (p = 0.03), increasing above the basal level at 7 days (p = 0.07). The bFGF basal level almost significantly decreased 2 days after infusion (p = 0.06), being at 7 days higher than the basal value (p = 0.09). Comparing the day 2 values with basal ones, the linear regression model showed a significant positive correlation between IL-8 and bFGF (p = 0.02), IL-8 and TNF-alpha (p < 0.0001), bFGF and TNF-alpha (p = 0.01), MMP-1 and TNF-alpha (p = 0.02). CONCLUSIONS ZA could exert an antiangiogenic activity and inhibition of tumor cell bone invasiveness by a transient reduction of VEGF, bFGF and MMP-2 circulating levels after infusion.
Collapse
Affiliation(s)
- Gianluigi Ferretti
- Division of Medical Oncology A, Regina Elena Cancer Institute, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Affiliation(s)
- Virginia Kaklamani
- Division of Hematology/Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | |
Collapse
|
46
|
Suarez-Cuervo C, Merrell MA, Watson L, Harris KW, Rosenthal EL, Väänänen HK, Selander KS. Breast cancer cells with inhibition of p38alpha have decreased MMP-9 activity and exhibit decreased bone metastasis in mice. Clin Exp Metastasis 2005; 21:525-33. [PMID: 15679050 DOI: 10.1007/s10585-004-3503-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
p38 belongs to a family of mitogen-activated protein kinases, which transfer extracellular signals into intracellular responses. p38 is also frequently detected in clinical breast cancer specimens, but its role as a prognostic factor is not known. Of the various p38 isoforms, p38alpha has been shown to mediate the in vitro invasiveness of breast cancer cells through up-regulation of urokinase plasminogen activator (uPA). We studied the role of p38alpha in breast cancer bone metastases, using dominant negative blockade approach. Human MDA-MB-231 breast cancer clones stably expressing dominant negative p38alpha (p38/AF) exhibited decreased basal MMP-9 activity. TGF-beta1-induced MMP-9 activity was also blunted in these clones, as compared with controls in which TGF-betal up-regulated MMP-9 activity. Consistent with these findings, SB202190, a specific p38 inhibitor, also inhibited TGF-beta1-induced MMP-9 activity in parental cells. The p38/AF clones exhibited also reduced uPA production after growth on vitronectin and decreased cell motility, as compared with controls. VEGF production levels in all the studied clones were similar. The p38/AF clone, which had similar in vitro growth rate as the control pcDNA3 clone, formed significantly less bone metastases in a mouse model, as compared with the control clone. In conclusion, inhibition of the p38alpha pathway results in decreased MMP-9 activity, impaired uPA expression and decreased motility, all of which may contribute to the decreased formation of bone metastasis.
Collapse
Affiliation(s)
- Catalina Suarez-Cuervo
- Department of Medicine, Division of Hematology-Oncology, University of Alabama at Birmingham, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Skrzydlewska E, Sulkowska M, Koda M, Sulkowski S. Proteolytic-antiproteolytic balance and its regulation in carcinogenesis. World J Gastroenterol 2005; 11:1251-66. [PMID: 15761961 PMCID: PMC4250670 DOI: 10.3748/wjg.v11.i9.1251] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer development is essentially a tissue remodeling process in which normal tissue is substituted with cancer tissue. A crucial role in this process is attributed to proteolytic degradation of the extracellular matrix (ECM). Degradation of ECM is initiated by proteases, secreted by different cell types, participating in tumor cell invasion and increased expression or activity of every known class of proteases (metallo-, serine-, aspartyl-, and cysteine) has been linked to malignancy and invasion of tumor cells. Proteolytic enzymes can act directly by degrading ECM or indirectly by activating other proteases, which then degrade the ECM. They act in a determined order, resulting from the order of their activation. When proteases exert their action on other proteases, the end result is a cascade leading to proteolysis. Presumable order of events in this complicated cascade is that aspartyl protease (cathepsin D) activates cysteine proteases (e.g., cathepsin B) that can activate pro-uPA. Then active uPA can convert plasminogen into plasmin. Cathepsin B as well as plasmin are capable of degrading several components of tumor stroma and may activate zymogens of matrix metalloproteinases, the main family of ECM degrading proteases. The activities of these proteases are regulated by a complex array of activators, inhibitors and cellular receptors. In physiological conditions the balance exists between proteases and their inhibitors. Proteolytic-antiproteolytic balance may be of major significance in the cancer development. One of the reasons for such a situation is enhanced generation of free radicals observed in many pathological states. Free radicals react with main cellular components like proteins and lipids and in this way modify proteolytic-antiproteolytic balance and enable penetration damaging cellular membrane. All these lead to enhancement of proteolysis and destruction of ECM proteins and in consequence to invasion and metastasis.
Collapse
Affiliation(s)
- Elzbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2, 15-230 Bialystok, Poland.
| | | | | | | |
Collapse
|
48
|
Morgan H, Hill PA. Human breast cancer cell-mediated bone collagen degradation requires plasminogen activation and matrix metalloproteinase activity. Cancer Cell Int 2005; 5:1. [PMID: 15701164 PMCID: PMC548674 DOI: 10.1186/1475-2867-5-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2004] [Accepted: 02/08/2005] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND: Breast cancer cells frequently metastasize to the skeleton and induce extensive bone destruction. Cancer cells produce proteinases, including matrix metalloproteinases (MMPs) and the plasminogen activator system (PAS) which promote invasion of extracellular matrices, but whether these proteinases degrade bone matrix is unclear. To characterize the role that breast cancer cell proteinases play in bone degradation we compared the effects of three human breast cancer cell lines, MDA-MB-231, ZR-75-1 and MCF-7 with those of a normal breast epithelial cell line, HME. The cell lines were cultured atop radiolabelled matrices of either mineralized or non-mineralized bone or type I collagen, the principal organic constituent of bone. RESULTS: The 3 breast cancer cell lines all produced significant degradation of the 3 collagenous extracellular matrices (ECMs) whilst the normal breast cell line was without effect. Breast cancer cells displayed an absolute requirement for serum to dissolve collagen. Degradation of collagen was abolished in plasminogen-depleted serum and could be restored by the addition of exogenous plasminogen. Localization of plasmin activity to the cell surface was critical for the degradation process as aprotinin, but not alpha2 antiplasmin, prevented collagen dissolution. During ECM degradation breast cancer cell lines expressed urokinase-type plasminogen activator (u-PA) and uPA receptor, and MMPs-1, -3, -9,-13, and -14. The normal breast epithelial cell line expressed low levels of MMPs-1, and -3, uPA and uPA receptor. Inhibitors of both the PAS (aprotinin and PA inhibitor-1) and MMPs (CT1166 and tisue inhibitor of metalloproteinase) blocked collagen degradation, demonstrating the requirement of both plasminogen activation and MMP activity for degradation. The activation of MMP-13 in human breast cancer cells was prevented by plasminogen activator inhibitor-1 but not by tissue inhibitor of metalloproteinase-1, suggesting that plasmin activates MMP-13 directly. CONCLUSIONS: These data demonstrate that breast cancer cells dissolve type I collagen and that there is an absolute requirement for plasminogen activation and MMP activity in the degradation process.
Collapse
Affiliation(s)
- Hayley Morgan
- Department of Craniofacial Biology and Orthodontics, King's College London, Floor 22, Guy's Tower, Guy's Hospital, London, SE1 9RT UK
| | - Peter A Hill
- Department of Craniofacial Biology and Orthodontics, King's College London, Floor 22, Guy's Tower, Guy's Hospital, London, SE1 9RT UK
| |
Collapse
|
49
|
Ilunga K, Nishiura R, Inada H, El-Karef A, Imanaka-Yoshida K, Sakakura T, Yoshida T. Co-stimulation of human breast cancer cells with transforming growth factor-beta and tenascin-C enhances matrix metalloproteinase-9 expression and cancer cell invasion. Int J Exp Pathol 2005; 85:373-9. [PMID: 15566434 PMCID: PMC2517534 DOI: 10.1111/j.0959-9673.2004.00406.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Transforming growth factor-beta (TGF-beta), tenascin-C (TN-C) and matrix metalloproteinases (MMPs) have been demonstrated independently to be associated with disease progression and poor prognosis in patients with breast cancer. The present study explored effects of TGF-beta and TN-C on MMP-9 expression and cancer invasion. An experimental study was designed to analyse MDA-MB-231 breast cancer cells, known for their high invasiveness, after stimulation with TGF-beta1 and/or TN-C. TGF-beta1 stimulated TN-C expression in the cells. Co-stimulation of MDA-MB-231 cells with TN-C and TGF-beta increased MMP-9 expression at both the gene (28-fold) and the protein levels. The in vitro invasion also increased (4-fold). GM6001 inhibited the invasion induced by the co-stimulation. The combined effect of TN-C and TGF-beta resulted in enhanced MMP-9 expression and cancer invasion in vitro.
Collapse
Affiliation(s)
- Kalembeyi Ilunga
- Department of Pathology, Mie University School of MedicineMie, Japan
- Department of Surgery, Lubumbashi University, Faculty of MedicineLubumbashi, Congo
| | - Rika Nishiura
- Department of Pathology, Mie University School of MedicineMie, Japan
| | - Hiroyasu Inada
- Department of Pathology, Mie University School of MedicineMie, Japan
| | - Amro El-Karef
- Department of Pathology, Mie University School of MedicineMie, Japan
| | | | - Teruyo Sakakura
- Department of Pathology, Mie University School of MedicineMie, Japan
| | | |
Collapse
|
50
|
Kim WU, Min SY, Cho ML, Hong KH, Shin YJ, Park SH, Cho CS. Elevated matrix metalloproteinase-9 in patients with systemic sclerosis. Arthritis Res Ther 2004; 7:R71-9. [PMID: 15642145 PMCID: PMC1064883 DOI: 10.1186/ar1454] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2004] [Revised: 09/23/2004] [Accepted: 10/01/2004] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) has been implicated in the pathogenesis of cancer, autoimmune disease, and various pathologic conditions characterized by excessive fibrosis. In this study, we investigated the expression of MMP-9 and its clinical significance in systemic sclerosis (SSc). The patients (n = 42) with SSc had higher concentrations of MMP-9 and of tissue inhibitor of metalloproteinase-1 (TIMP-1) and a higher ratio of MMP-9 to TIMP-1 in sera than healthy controls (n = 32). Serum MMP-9 concentrations were significantly higher in the diffuse type (n = 23) than the limited type of SSc (n = 19). Serum concentrations of MMP-9 correlated well with the degree of skin involvement, as determined by the Rodnan score and with serum concentrations of transforming growth factor β. Moreover, dermal fibroblasts from patients with SSc produced more MMP-9 than those from healthy controls when they were stimulated with IL-1β, tumor necrosis factor α, or transforming growth factor β. Such an increase in MMP-9 production was partially blocked by treatment with cyclosporin A. In summary, the serum MMP-9 concentrations were elevated in SSc patients and correlated well with skin scores. The increased MMP-9 concentrations may be attributable to overproduction by dermal fibroblasts in SSc. These findings suggest that the enhanced production of MMP-9 may contribute to fibrogenic remodeling during the progression of skin sclerosis in SSc.
Collapse
Affiliation(s)
- Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, Catholic Research Institutes of Medical Science, School of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|