1
|
Tanabe H, Suzuki T, Ohishi T, Isemura M, Nakamura Y, Unno K. Effects of Epigallocatechin-3-Gallate on Matrix Metalloproteinases in Terms of Its Anticancer Activity. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020525. [PMID: 36677584 PMCID: PMC9862901 DOI: 10.3390/molecules28020525] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023]
Abstract
Epidemiological studies have shown that the consumption of green tea has beneficial effects against cancer. Basic studies have provided evidence that epigallocatechin gallate (EGCG) is a major contributor to these effects. Matrix metalloproteinases (MMPs) are zinc-dependent metalloproteinases with the ability to degrade the extracellular matrix proteins and are involved in various diseases including cancer in which MMPs have a critical role in invasion and metastasis. In this review, we discuss the effects of EGCG on several types of MMPs in the context of its anticancer activity. In the promoter region, MMPs have binding sites for at least one transcription factor of AP-1, Sp1, and NF-κB, and EGCG can downregulate these transcription factors through signaling pathways mediated by reactive oxygen species. EGCG can also decrease nuclear ERK, p38, heat shock protein-27 (Hsp27), and β-catenin levels, leading to suppression of MMPs' expression. Other mechanisms by which EGCG inhibits MMPs include direct binding to MMPs to prevent their activation and downregulation of NF-κB to suppress the production of inflammatory cytokines such as TNFα and IL-1β. Findings from studies on EGCG presented here may be useful in the development of more effective anti-MMP agents, which would give beneficial effects on cancer and other diseases.
Collapse
Affiliation(s)
- Hiroki Tanabe
- Faculty of Health and Welfare Science, Nayoro City University, Nayoro 096-8641, Hokkaido, Japan
- Correspondence: (H.T.); (T.O.)
| | - Takuji Suzuki
- Department of Food Science and Nutrition, Faculty of Human Life and Science, Doshisha Women’s College of Liberal Arts, Kyoto 602-0893, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu 410-0301, Shizuoka, Japan
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, Shinagawa, Tokyo 141-0021, Japan
- Correspondence: (H.T.); (T.O.)
| | - Mamoru Isemura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yoriyuki Nakamura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Keiko Unno
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
2
|
Wu N, Liu GB, Zhang YM, Wang Y, Zeng HT, Xiang H. MiR-708-5p/Pit-1 axis mediates high phosphate-induced calcification in vascular smooth muscle cells via Wnt8b/β-catenin pathway. Kaohsiung J Med Sci 2022; 38:653-661. [PMID: 35460325 DOI: 10.1002/kjm2.12542] [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: 05/13/2021] [Revised: 01/10/2022] [Accepted: 03/01/2022] [Indexed: 11/06/2022] Open
Abstract
Recently, the underlying mechanism of vascular calcification (VC) has been partially elucidated. However, it is still high incidence, and no effective treatment has been found. This study aims at figuring out the underlying mechanisms of microRNA-708-5p (miR-708-5p)/sodium-phosphate transporter 1 (Pit-1) axis in high phosphate (HP)-induced VC of T/G HA-VSMCs. Alizarin Red S staining was used to evaluate calcium salt deposition, and the activity of alkaline phosphatase (ALP) was determined by measuring the absorbance at 405 nm. RT-qPCR and Western blot were performed to assess the levels of miR-708-5p and Pit-1, the levels of ALP, Pit-1, β-catenin, glycogen synthesis kinase 3 β (GSK3β), and p-GSK3β proteins, respectively. The interaction between miR-708-5p and Pit-1 was validated by luciferase reporter assay. Our findings illustrated that miR-708-5p was downregulated and Pit-1was upregulated in HP-induced VC. MiR-708-5p mimics inhibited HP-induced VC. Further experiments demonstrated that miR-708-5p targets Pit-1. In addition, miR-708-5p inactivates the Wnt8b/β-catenin pathway via targeting Pit-1 to reduce HP-induced VC. MiR-708-5p has a crucial effect on VC via targeting Pit-1 and inhibiting Wnt8b/β-catenin pathway, it may serve as a new target for VC treatment.
Collapse
Affiliation(s)
- Na Wu
- Department of Cardiology, The Third Hospital of Changsha, Changsha, China
| | - Guo-Bing Liu
- Department of Cardiology, The Third Hospital of Changsha, Changsha, China
| | - Yu-Min Zhang
- Department of Cardiology, The Third Hospital of Changsha, Changsha, China
| | - Yong Wang
- Department of Cardiology, The Third Hospital of Changsha, Changsha, China
| | - Hai-Tao Zeng
- Department of Cardiology, The Third Hospital of Changsha, Changsha, China
| | - Hui Xiang
- Department of Nephrology, The Third Hospital of Changsha, Changsha, China
| |
Collapse
|
3
|
Sauchinone inhibits the proliferation, migration and invasion of breast cancer cells by suppressing Akt-CREB-MMP13 signaling pathway. Biosci Rep 2021; 41:229926. [PMID: 34643237 PMCID: PMC8561391 DOI: 10.1042/bsr20211067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/01/2021] [Accepted: 10/12/2021] [Indexed: 11/17/2022] Open
Abstract
Sauchinone, a lignan isolated from Saururus chinenesis, is known to exhibit anti-inflammatory and anti-oxidant effects. Recently, sauchinone has been reported to inhibit the growth of various cancer cells, but its effects on breast cancer cells remain poorly understood. In the present study, we investigated the effects of sauchinone on the growth of breast cancer cells along with the underlying molecular mechanisms. Our results show that sauchinone treatment markedly inhibited the proliferation, migration, and invasion of breast cancer cells. Sauchinone reduced the phosphorylation of Akt, ERK, and CREB increased by transforming growth factor-β (TGF-β). In particular, sauchinone treatment suppressed the expression of matrix metalloproteinase (MMP)-13 (MMP13) by regulating the Akt-CREB signaling pathway. Sauchinone was less effective in inhibiting cell migration in Mmp13-knockdown cells than in control cells, suggesting that MMP13 may be a novel target for sauchinone. Our study suggests that sauchinone inhibits the growth of breast cancer cells by attenuating the Akt-CREB-MMP13 pathway. In addition, the targeted inhibition of MMP13 by sauchinone represents a promising approach for the treatment of breast cancer.
Collapse
|
4
|
Xu K, Zhang W, Wang C, Hu L, Wang R, Wang C, Tang L, Zhou G, Zou B, Xie H, Tang J, Guan X. Integrative analyses of scRNA-seq and scATAC-seq reveal CXCL14 as a key regulator of lymph node metastasis in breast cancer. Hum Mol Genet 2021; 30:370-380. [PMID: 33564857 DOI: 10.1093/hmg/ddab042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 01/05/2023] Open
Abstract
The potentially different genetics and epigenetics in the primary tumors and metastases affect the efficacy of treatment in breast cancer patients. Nevertheless, the cellular and molecular mechanisms of breast cancer lymph node metastasis still remain elusive. Here, we employed single-cell RNA sequencing to acquire the transcriptomic profiles of individual cells from primary tumors, negative lymph nodes (NLs) and positive lymph nodes (PLs). We also performed a single-cell assay for transposase-accessible chromatin (ATAC) sequencing (scATAC-seq) of the positive and NL samples to get the chromatin accessibility profile. We identified a novel cell subpopulation with an abnormally high expression level of CXCL14 in the PL of breast cancer patients. Cell trajectory analysis also revealed that CXCL14 was increased expressed in the late pseudo-time. Moreover, based on a tissue microarray of 55 patients and the Oncomine database, we validated that CXCL14 expression was significantly higher in breast cancer patients with lymph node metastasis. Furthermore, scATAC-seq identified several transcription factors that may be potential regulation factors for the lymph node metastasis of breast cancer. Thus, our findings will improve our current understanding of the mechanism for lymph node metastasis, and they are potentially valuable in providing novel prognosis markers for the lymphatic metastasis of breast cancer.
Collapse
Affiliation(s)
- Kun Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Wenwen Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Cong Wang
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Longfei Hu
- Singleron Biotechnologies, Yaogu Avenue 11, Nanjing 210061, Jiangsu, China
| | - Runtian Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Cenzhu Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Lin Tang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Guohua Zhou
- Department of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Bingjie Zou
- Department of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Hui Xie
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jinhai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
5
|
Sung NJ, Kim NH, Surh YJ, Park SA. Gremlin-1 Promotes Metastasis of Breast Cancer Cells by Activating STAT3-MMP13 Signaling Pathway. Int J Mol Sci 2020; 21:ijms21239227. [PMID: 33287358 PMCID: PMC7730512 DOI: 10.3390/ijms21239227] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
Gremlin-1 (GREM1), one of the bone morphogenetic protein (BMP) antagonists, can directly bind to BMPs. GREM1 is involved in organogenesis, tissue differentiation, and organ fibrosis. Recently, numerous studies have reported the oncogenic role of GREM1 in cancer. However, the role of GREM1 in metastasis of breast cancer cells and its underlying mechanisms remain poorly understood. The role of GREM1 in breast cancer progression was assessed by measuring growth, migration, and invasion of breast cancer cells. An orthotopic breast cancer mouse model was used to investigate the role of GREM1 in lung metastasis of breast cancer cells. GREM1 knockdown suppressed the proliferation of breast cancer cells, while its overexpression increased their growth, migration, and invasion. Cells with Grem1-knockdown showed much lower tumor growth rates and lung metastasis than control cells. GREM1 enhanced the expression of matrix metalloproteinase 13 (MMP13). A positive correlation between GREM1 and MMP13 expression was observed in breast cancer patients. GREM1 activated signal transducer and activator of transcription 3 (STAT3) transcription factor involved in the expression of MMP13. Our study suggests that GREM1 can promote lung metastasis of breast cancer cells through the STAT3-MMP13 pathway. In addition, GREM1 might be a promising therapeutic target for breast cancer metastasis.
Collapse
Affiliation(s)
- Nam Ji Sung
- Department of ICT Environmental Health System, Graduate School, Soonchunhyang University, Asan-si 31538, Korea; (N.J.S.); (N.H.K.)
| | - Na Hui Kim
- Department of ICT Environmental Health System, Graduate School, Soonchunhyang University, Asan-si 31538, Korea; (N.J.S.); (N.H.K.)
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, Korea;
| | - Sin-Aye Park
- Department of ICT Environmental Health System, Graduate School, Soonchunhyang University, Asan-si 31538, Korea; (N.J.S.); (N.H.K.)
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: ; Tel.: +82-41-530-4990
| |
Collapse
|
6
|
Seoane S, Martinez-Ordoñez A, Eiro N, Cabezas-Sainz P, Garcia-Caballero L, Gonzalez LO, Macia M, Sanchez L, Vizoso F, Perez-Fernandez R. POU1F1 transcription factor promotes breast cancer metastasis via recruitment and polarization of macrophages. J Pathol 2019; 249:381-394. [PMID: 31292963 DOI: 10.1002/path.5324] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/21/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022]
Abstract
Cancer progression requires cells surrounding tumors be reeducated and activated to support tumor growth. Oncogenic signals from malignant cells directly influence stromal composition and activation, but the factors mediating this communication are still not well understood. We have previously shown that the transcription factor POU class 1 homeobox 1 (POU1F1), also known as Pit-1, induces profound changes on neoplastic cell-autonomous processes favoring metastasis in human breast cancer. Here we describe for the first time Pit-1-mediated paracrine actions on macrophages in the tumor microenvironment by using cell lines in vitro, zebrafish and mouse models in vivo, and samples from human breast cancer patients. Through the release of CXCL12, Pit-1 in tumor cells was found to mediate the recruitment and polarization of macrophages into tumor-associated macrophages (TAMs). In turn, TAMs collaborated with tumor cells to increase tumor growth, angiogenesis, extravasation and metastasis to lung. Our data reveal a new mechanism of cooperation between tumor cells and macrophages favoring metastasis and poor clinical outcome in human breast cancer, which suggests that Pit-1 and CXCL12 should be further studied as potential prognostic and therapeutic indicators. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Samuel Seoane
- Department of Physiology - Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Anxo Martinez-Ordoñez
- Department of Physiology - Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Noemi Eiro
- Research Unit, Hospital Fundacion de Jove, Gijón, Spain
| | - Pablo Cabezas-Sainz
- Department of Zoology, Genetics and Physical Anthropology, University of Santiago de Compostela, Lugo, Spain
| | - Lucia Garcia-Caballero
- Department of Morphological Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Manuel Macia
- Department of Obstetrics and Gynecology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Laura Sanchez
- Department of Zoology, Genetics and Physical Anthropology, University of Santiago de Compostela, Lugo, Spain
| | | | - Roman Perez-Fernandez
- Department of Physiology - Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
7
|
Sendon-Lago J, Seoane S, Martinez-Ordoñez A, Eiro N, Saa J, Vizoso FJ, Gonzalez F, Perez-Fernandez R, Bermudez MA. Corneal regeneration by conditioned medium of human uterine cervical stem cells is mediated by TIMP-1 and TIMP-2. Exp Eye Res 2019; 180:110-121. [PMID: 30557571 DOI: 10.1016/j.exer.2018.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/22/2018] [Accepted: 12/11/2018] [Indexed: 01/15/2023]
Abstract
The aim of the present study was to evaluate the effect and the mechanism of action of the conditioned medium from human uterine cervical stem cells (CM-hUCESC) on corneal wound healing in a rabbit dry eye model. To do this, dry eye and corneal epithelial injuries were induced in rabbits by topical administration of atropine sulfate and NaOH. Hematoxylin-Eosin (H&E) and Ki-67 immunostaining were carried out to evaluate corneal damage and cell proliferation, and real-time PCR was used to evaluate proinflammatory cytokines in the cornea. In addition, in order to investigate possible factors involved in corneal regeneration, primary cultures of rat corneal epithelial cells (rCECs) were used to evaluate cell migration, proliferation, and apoptosis before and after immunoprecipitation of specific factors from the CM-hUCESC. Results showed that CM-hUCESC treatment significantly improved epithelial regeneration in rabbits with dry eye induced by atropine and reduced corneal pro-inflammatory TNF-α, MCP-1, MIP-1α and IL-6 cytokines. In addition, metalloproteinase inhibitors TIMP-1 and TIMP-2, which are present at high levels in CM-hUCESC, mediated corneal regenerative effects by both inducing corneal epithelial cell proliferation and inhibiting apoptosis. In summary, CM-hUCESC induces faster corneal regeneration in a rabbit model of dry eye induced by atropine than conventional treatments, being TIMP-1 and TIMP-2 mediators in this process. The results indicate that an alternative CM-based treatment for some corneal conditions is achievable, although future studies would be necessary to investigate other factors involved in the multiple observed effects of CM-hUCESC.
Collapse
Affiliation(s)
- Juan Sendon-Lago
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Samuel Seoane
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Anxo Martinez-Ordoñez
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Noemi Eiro
- Research Unit, Hospital Fundacion de Jove, Gijón, Spain.
| | - Jorge Saa
- Research Unit, Hospital Fundacion de Jove, Gijón, Spain; Service of Ophthalmology, Fundación Hospital de Jove, Gijón, Spain.
| | | | - Francisco Gonzalez
- Department of Surgery and CIMUS, University of Santiago de Compostela, Spain; Service of Ophthalmology and IDIS, Complejo Hospitalario Universitario de Santiago de Compostela, Spain.
| | - Roman Perez-Fernandez
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Maria A Bermudez
- Department of Biology, Faculty of Science, University of A Coruña, Spain.
| |
Collapse
|
8
|
Wang H, Huang Y, Shi J, Zhi Y, Yuan F, Yu J, Chen Z, Yang J. XPC deficiency leads to centrosome amplification by inhibiting BRCA1 expression upon cisplatin-mediated DNA damage in human bladder cancer. Cancer Lett 2018; 444:136-146. [PMID: 30579971 DOI: 10.1016/j.canlet.2018.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/03/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
Xeroderma pigmentosum group C (XPC) is a well-known DNA damage recognition protein. Defects in XPC lead to carcinogenesis and progression of many human cancers. In the current study, we defined a novel, important role of XPC in preventing centrosome amplification during cisplatin-mediated DNA damage response. From experiments with human bladder cancer tissue, urothelial tissue from Xpc knockout mice and XPC-silenced cell lines, we found that attenuated XPC expression was associated with increased centrosome amplification in human bladder cancer. A significant increase in centrosome amplification was observed in XPC-silenced cells upon cisplatin treatment. XPC deficiency leads to reduced BRCA1 expression via upregulating its transcriptional repressor, Pit-1. The BRCA1 downregulation results in more DNA double strand breaks accumulation and persistent activation of the ATM-Chk1/Chk2 signaling, resulting in a prolonged G2/M arrest during which centrosome can over-duplicate and lead to centrosome amplification. XPC complementation in silenced cells could reduce Pit-1 expression, increase BRCA1 expression and recover the status of centrosome amplification. Our study reveals a new function for XPC in preventing chromosomal instability, providing new information on cancer chemotherapy and potential clinical significance for cancer management.
Collapse
Affiliation(s)
- Huanhuan Wang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Yaqin Huang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Jiazhong Shi
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Yi Zhi
- Department of Urology, Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Fang Yuan
- Chongqing University Cancer Hospital, Chongqing, PR China
| | - Jin Yu
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Zhiwen Chen
- Urology Institute of People's Liberation Army, Southwest Hospital, The Third Military Medical University, Chongqing, PR China; Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.
| | - Jin Yang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China.
| |
Collapse
|
9
|
Dumortier M, Ladam F, Damour I, Vacher S, Bièche I, Marchand N, de Launoit Y, Tulasne D, Chotteau-Lelièvre A. ETV4 transcription factor and MMP13 metalloprotease are interplaying actors of breast tumorigenesis. Breast Cancer Res 2018; 20:73. [PMID: 29996935 PMCID: PMC6042225 DOI: 10.1186/s13058-018-0992-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 05/23/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The ETS transcription factor ETV4 is involved in the main steps of organogenesis and is also a significant mediator of tumorigenesis and metastasis, such as in breast cancer. Indeed, ETV4 is overexpressed in breast tumors and is associated with distant metastasis and poor prognosis. However, the cellular and molecular events regulated by this factor are still misunderstood. In mammary epithelial cells, ETV4 controls the expression of many genes, MMP13 among them. The aim of this study was to understand the function of MMP13 during ETV4-driven tumorigenesis. METHODS Different constructs of the MMP13 gene promoter were used to study the direct regulation of MMP13 by ETV4. Moreover, cell proliferation, migration, invasion, anchorage-independent growth, and in vivo tumorigenicity were assayed using models of mammary epithelial and cancer cells in which the expression of MMP13 and/or ETV4 is modulated. Importantly, the expression of MMP13 and ETV4 messenger RNA was characterized in 456 breast cancer samples. RESULTS Our results revealed that ETV4 promotes proliferation, migration, invasion, and anchorage-independent growth of the MMT mouse mammary tumorigenic cell line. By investigating molecular events downstream of ETV4, we found that MMP13, an extracellular metalloprotease, was an ETV4 target gene. By overexpressing or repressing MMP13, we showed that this metalloprotease contributes to proliferation, migration, and anchorage-independent clonogenicity. Furthermore, we demonstrated that MMP13 inhibition disturbs proliferation, migration, and invasion induced by ETV4 and participates to ETV4-induced tumor formation in immunodeficient mice. Finally, ETV4 and MMP13 co-overexpression is associated with poor prognosis in breast cancer. CONCLUSION MMP13 potentiates the effects of the ETV4 oncogene during breast cancer genesis and progression.
Collapse
Affiliation(s)
- Mandy Dumortier
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000, Lille, France
| | - Franck Ladam
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605-2324, USA
| | - Isabelle Damour
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000, Lille, France
| | - Sophie Vacher
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | - Ivan Bièche
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | - Nathalie Marchand
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000, Lille, France
| | - Yvan de Launoit
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000, Lille, France
| | - David Tulasne
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000, Lille, France
| | - Anne Chotteau-Lelièvre
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000, Lille, France. .,CNRS UMR 8161, Institut de Biologie de Lille - Institut Pasteur de Lille, 1 Rue Pr Calmette, BP447, 59021, Lille, France.
| |
Collapse
|
10
|
Martinez-Ordoñez A, Seoane S, Cabezas P, Eiro N, Sendon-Lago J, Macia M, Garcia-Caballero T, Gonzalez LO, Sanchez L, Vizoso F, Perez-Fernandez R. Breast cancer metastasis to liver and lung is facilitated by Pit-1-CXCL12-CXCR4 axis. Oncogene 2018; 37:1430-1444. [PMID: 29321662 DOI: 10.1038/s41388-017-0036-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/13/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023]
Abstract
Development of human tumors is driven by accumulation of alterations in tumor suppressor genes and oncogenes in cells. The POU1F1 transcription factor (also known Pit-1) is expressed in the mammary gland and its overexpression induces profound phenotypic changes in proteins involved in breast cancer progression. Patients with breast cancer and elevated expression of Pit-1 show a positive correlation with the occurrence of distant metastasis and poor overall survival. However, some mediators of Pit-1 actions are still unknown. Here, we show that CXCR4 chemokine receptor and its ligand CXCL12 play a critical role in the pro-tumoral process induced by Pit-1. We found that Pit-1 increases mRNA and protein in both CXCR4 and CXCL12. Knock-down of CXCR4 reduces tumor growth and spread of Pit-1 overexpressing cells in a zebrafish xenograft model. Furthermore, we described for the first time pro-angiogenic effects of Pit-1 through the CXCL12-CXCR4 axis, and that extravasation of Pit-1 overexpressing breast cancer cells is strongly reduced in CXCL12-deprived target tissues. Finally, in breast cancer patients, expression of Pit-1 in primary tumors was found to be positively correlated with CXCR4 and CXCL12, with specific metastasis in liver and lung, and with clinical outcome. Our results suggest that Pit-1-CXCL12-CXCR4 axis could be involved in chemotaxis guidance during the metastatic process, and may represent prognostic and/or therapeutic targets in breast tumors.
Collapse
Affiliation(s)
- Anxo Martinez-Ordoñez
- Department of Physiology-Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Samuel Seoane
- Department of Physiology-Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Pablo Cabezas
- Department of Genetics, University of Santiago de Compostela, Campus de Lugo, Lugo, Spain
| | - Noemi Eiro
- Research Unit, Hospital Fundacion de Jove, Gijón, Spain
| | - Juan Sendon-Lago
- Department of Physiology-Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Macia
- Department of Obstetrics and Gynecology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Tomas Garcia-Caballero
- Department of Morphological Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Laura Sanchez
- Department of Genetics, University of Santiago de Compostela, Campus de Lugo, Lugo, Spain
| | | | - Roman Perez-Fernandez
- Department of Physiology-Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
11
|
Sendon-Lago J, Seoane S, Eiro N, Bermudez MA, Macia M, Garcia-Caballero T, Vizoso FJ, Perez-Fernandez R. Erratum to: Cancer progression by breast tumors with Pit-1-overexpression is blocked by inhibition of metalloproteinase (MMP)-13. Breast Cancer Res 2017; 19:38. [PMID: 28351385 PMCID: PMC5369202 DOI: 10.1186/s13058-017-0834-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 11/10/2022] Open
Affiliation(s)
- Juan Sendon-Lago
- Department of Physiology- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), School of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Samuel Seoane
- Department of Physiology- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), School of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Noemi Eiro
- Unidad de Investigación, Fundacion Hospital de Jove, 33290, Gijón, Spain
| | - Maria A Bermudez
- Department of Physiology- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), School of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Manuel Macia
- Departments of Obstetrics and Gynecology, School of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Tomas Garcia-Caballero
- Departments of Morphological Sciences, School of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Francisco J Vizoso
- Unidad de Investigación, Fundacion Hospital de Jove, 33290, Gijón, Spain
| | - Roman Perez-Fernandez
- Department of Physiology- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), School of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
12
|
Wang O, Yang F, Liu Y, Lv L, Ma R, Chen C, Wang J, Tan Q, Cheng Y, Xia E, Chen Y, Zhang X. C-MYC-induced upregulation of lncRNA SNHG12 regulates cell proliferation, apoptosis and migration in triple-negative breast cancer. Am J Transl Res 2017; 9:533-545. [PMID: 28337281 PMCID: PMC5340688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/05/2017] [Indexed: 06/06/2023]
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer, with a significantly higher recurrence and mortality rate. There is an urgent need to uncover the mechanism underlying TNBC and establish therapeutic targets. Long non-coding RNAs (lncRNAs) are involved in a series of biological functions and provide novel insights into the molecular mechanism of cancer. Based on their expression specificity and large number, lncRNAs are likely to serve as the basis for clinical applications in oncology. In our previous study, we utilized RNA sequencing (RNA-seq) to explore the lncRNAs expression profiles in TNBC and identified that small nucleolar RNA host gene 12 (SNHG12) was remarkably increased in TNBC. However, the role of SNHG12 in TNBC has not been clarified. Herein, we determine that SNHG12 is upregulated in TNBC, and its high expression is significantly correlated with tumor size and lymph node metastasis. Mechanistic investigations show that SNHG12 is a direct transcriptional target of c-MYC. Silencing SNHG12 expression inhibits TNBC cells proliferation and apoptosis promotion, whereas SNHG12 overexpression has the opposite effect. In addition, we reveal that SNHG12 may promote cells migration by regulating MMP13 expression. To the best of our knowledge, it is the first report indicating that SNHG12 is involved in breast cancer. Taken together, our findings suggest that SNHG12 contributes to the oncogenic potential of TNBC and may be a promising therapeutic target.
Collapse
Affiliation(s)
- Ouchen Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Fan Yang
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Yehuan Liu
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Lin Lv
- Department of Oncology, Jinhua Municipal Central HospitalJinhua, Zhejiang, PR China
| | - Ruimin Ma
- Department of Breast Surgery, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Chuanzhi Chen
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Jiao Wang
- Department of Eye Hospital, Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Qiufan Tan
- Department of Eye Hospital, Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Yue Cheng
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Erjie Xia
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Yizuo Chen
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| | - Xiaohua Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, Zhejiang, PR China
| |
Collapse
|
13
|
Vitamin D and the Epithelial to Mesenchymal Transition. Stem Cells Int 2016; 2016:6213872. [PMID: 26880977 PMCID: PMC4736588 DOI: 10.1155/2016/6213872] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/08/2015] [Indexed: 12/13/2022] Open
Abstract
Several studies support reciprocal regulation between the active vitamin D derivative 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) and the epithelial to mesenchymal transition (EMT). Thus, 1,25(OH)2D3 inhibits EMT via the induction of a variety of target genes that encode cell adhesion and polarity proteins responsible for the epithelial phenotype and through the repression of key EMT inducers. Both direct and indirect regulatory mechanisms mediate these effects. Conversely, certain master EMT inducers inhibit 1,25(OH)2D3 action by repressing the transcription of VDR gene encoding the high affinity vitamin D receptor that mediates 1,25(OH)2D3 effects. Consequently, the balance between the strength of 1,25(OH)2D3 signaling and the induction of EMT defines the cellular phenotype in each context. Here we review the current understanding of the genes and mechanisms involved in the interplay between 1,25(OH)2D3 and EMT.
Collapse
|
14
|
Chen JH, Tsai CH, Lin HY, Huang CF, Leung YM, Lai SW, Tsai CF, Chang PC, Lu DY, Lin C. Interlukin-18 Is a Pivot Regulatory Factor on Matrix Metalloproteinase-13 Expression and Brain Astrocytic Migration. Mol Neurobiol 2015; 53:6218-6227. [PMID: 26558633 DOI: 10.1007/s12035-015-9529-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/05/2015] [Indexed: 01/28/2023]
Abstract
The expression of matrix metalloproteinase-13 (MMP-13) has been shown to be elevated in some pathophysiological conditions and is involved in the degradation of extracellular matrix in astrocytes. In current study, the function of MMP-13 was further investigated. The conditioned medium (CM) collected from activated microglia increased interleukin (IL)-18 production and enhanced MMP-13 expression in astrocytes. Furthermore, treatment with recombinant IL-18 increased MMP-13 protein and mRNA levels in astrocytes. Recombinant IL-18 stimulation also increased the enzymatic activity of MMP-13 and the migratory activity of astrocytes, while administration of MMP-13 or pan-MMP inhibitors antagonized IL-18-induced migratory activity of astrocytes. In addition, administration of recombinant IL-18 to astrocytes led to the phosphorylation of JNK, Akt, or PKCδ, and treatment of astrocytes with JNK, PI3 kinase/Akt, or PKCδ inhibitors significantly decreased the IL-18-induced migratory activity. Taken together, the results suggest that IL-18-induced MMP-13 expression in astrocytes is regulated by JNK, PI3 kinase/Akt, and PKCδ signaling pathways. These findings also indicate that IL-18 is an important regulator leading to MMP-13 expression and cell migration in astrocytes.
Collapse
Affiliation(s)
- Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chon-Haw Tsai
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Hsiao-Yun Lin
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Chien-Fang Huang
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Yuk-Man Leung
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Sheng-Wei Lai
- Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics, Asia University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan. .,Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan.
| |
Collapse
|
15
|
Artacho-Cordón F, Ríos-Arrabal S, Olivares-Urbano MA, Storch K, Dickreuter E, Muñoz-Gámez JA, León J, Calvente I, Torné P, Salinas MDM, Cordes N, Núñez MI. Valproic acid modulates radiation-enhanced matrix metalloproteinase activity and invasion of breast cancer cells. Int J Radiat Biol 2015; 91:946-56. [PMID: 26490761 DOI: 10.3109/09553002.2015.1087067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To evaluate matrix metalloproteinase (MMP) activity and invasion after ionizing radiation (IR) exposure and to determine whether MMP could be epigenetically modulated by histone deacetylase (HDAC) inhibition. MATERIAL AND METHODS Two human breast cancer cell lines (MDA-MB-231 and MCF-7) were cultured in monolayer (2D) and in laminin-rich extracellular matrix (3D). Invasion capability, collagenolytic and gelatinolytic activity, MMP and TIMP protein and mRNA expression and clonogenic survival were analyzed after IR exposure, with and without a HDAC inhibition treatment [1.5 mM valproic acid (VA) or 1 μM trichostatin-A (TSA)]. RESULTS IR exposure resulted in cell line-dependent stimulation of invasion capacity. In contrast to MCF-7 cells, irradiated MDA-MB-231 showed significantly enhanced mRNA expression of mmp-1, mmp-3 and mmp-13 and of their regulators timp-1 and timp-2 relative to unirradiated controls. This translated into increased collagenolytic and gelatinolytic activity and could be reduced after valproic acid (VA) treatment. Additionally, VA also mitigated IR-enhanced mmp and timp mRNA expression as well as IR-increased invasion capability. Finally, our data confirm the radiosensitizing effect of VA. CONCLUSION These results suggest that IR cell line-dependently induces upregulation of MMP mRNA expression, which appears to be mechanistically linked to a higher invasion capability that is modifiable by HDAC inhibition.
Collapse
Affiliation(s)
- Francisco Artacho-Cordón
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain
| | - Sandra Ríos-Arrabal
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,c Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada , Armilla, Granada , Spain
| | | | - Katja Storch
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - Ellen Dickreuter
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - José Antonio Muñoz-Gámez
- b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,i CIBER on hepatic and digestive diseases (CIBEREHD) , Spain
| | - Josefa León
- b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,i CIBER on hepatic and digestive diseases (CIBEREHD) , Spain
| | - Irene Calvente
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain
| | - Pablo Torné
- j General Surgery Management Unit, San Cecilio University Hospital , Granada , Spain
| | - María del Mar Salinas
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain
| | - Nils Cordes
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - María Isabel Núñez
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,c Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada , Armilla, Granada , Spain
| |
Collapse
|