1
|
Sasi N, Preetha D, Iyyappan S, Selvamurugan N. Circular RNAs: Emerging regulators of signaling pathways in epithelial-mesenchymal transition and angiogenesis during breast cancer progression. Semin Oncol 2025; 52:152340. [PMID: 40220369 DOI: 10.1016/j.seminoncol.2025.152340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 04/14/2025]
Abstract
Circular RNAs (circRNAs) have emerged as important regulators of gene expression and cellular activities, and abnormalities in circRNAs in breast cancer have been linked to important biological processes like epithelial-mesenchymal transition (EMT) and angiogenesis, both essential for tumor metastasis. EMT facilitates the transition of epithelial cancer cells into a mesenchymal phenotype, enhancing their invasive and migratory capabilities, while angiogenesis promotes tumor progression by forming new blood vessels. CircRNAs also interact with microRNAs to regulate signaling pathways such as TGF-β, Wnt/-catenin, and VEGF. Besides EMT and angiogenesis, studies have identified that circRNAs affect metabolic reprogramming, chemoresistance, tumor microenvironment remodeling, and immunological evasion. Thus, circRNAs play a multifaceted role in the development of breast cancer. They hold potential as non-invasive biomarkers and therapeutic targets due to their high stability, resistance to exonuclease degradation, abundance in body fluids, and diverse expression patterns across different tissues. This review summarizes and critically assesses existing understanding of the functional roles and molecular processes of circRNAs in controlling EMT and angiogenesis during breast cancer progression.
Collapse
Affiliation(s)
- Nivruthi Sasi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, India
| | - Dilipkumar Preetha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, India
| | - Saranya Iyyappan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, India.
| |
Collapse
|
2
|
Wen P, Jiang D, Qu F, Wang G, Zhang N, Shao Q, Huang Y, Li S, Wang L, Zeng X. PFDN5 plays a dual role in breast cancer and regulates tumor immune microenvironment: Insights from integrated bioinformatics analysis and experimental validation. Gene 2025; 933:149000. [PMID: 39396557 DOI: 10.1016/j.gene.2024.149000] [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: 05/08/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Although the prognosis for patients with breast cancer has improved, breast cancer remains the leading cause of death for women worldwide. Prefoldin 5 (PFDN5), as a subunit of the prefoldin complex, plays a vital role in aiding the correct folding of newly synthesized proteins. However, the exact impact of PFDN5 on breast cancer development and its prognostic implications remain unclear. METHODS We conducted bioinformatics analysis to investigate the correlation between PFDN5 and patient survival, as well as various clinicopathological characteristics in breast cancer. Additionally, various assays were employed to validate the biological functions of PFDN5 in breast cancer. Finally, RNA sequencing (RNA-seq) was utilized to investigate the molecular mechanisms associated with PFDN5. RESULTS Compared to normal tissues, PFDN5 exhibited lower expression levels in breast cancer tissues, and lower expression of PFDN5 is associated with poorer prognosis. PFDN5 led to G2/M phase arrest in the cell cycle and reduced proliferative potential in breast cancer cells. However, PFDN5 also promoted migration and invasion of breast cancer cells. Also, RNA-seq analysis revealed an involvement of PFDN5 in the cell cycle and TGF-β signaling pathway. Furthermore, PFDN5 had a significant impact on tumor immune microenvironment by promoting macrophage polarization towards the M1 phenotype and exhibited a positive correlation with CD8+ T cell infiltration levels. CONCLUSIONS PFDN5 plays a dual role in breast cancer and serves as a key factor in tumor immune microenvironment. Therefore, PFDN5 holds promise as a valuable biomarker for predicting both metastatic and prognosis in breast cancer.
Collapse
Affiliation(s)
- Ping Wen
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Dongping Jiang
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Fanli Qu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Guanwen Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Ningning Zhang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Qing Shao
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yuxin Huang
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Sisi Li
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Long Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China.
| | - Xiaohua Zeng
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China; Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing 400030, China.
| |
Collapse
|
3
|
Giacobbi E, Bonfiglio R, Rotondaro G, Servadei F, Smirnov A, Palumbo V, Scioli MP, Bonanno E, Buonomo CO, Vanni G, Candi E, Mauriello A, Scimeca M. Implications of Mineralization Biomarkers in Breast Cancer Outcomes Beyond Calcifications. Int J Mol Sci 2025; 26:645. [PMID: 39859358 PMCID: PMC11765781 DOI: 10.3390/ijms26020645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/30/2025] Open
Abstract
The aim of this work was to explore the biomarkers associated with epithelial to mesenchymal transition (EMT) and mineralization processes as new prognostic factors across different breast cancer phenotypes. To this end, 133 breast biopsies, including benign and malignant lesions, with or without microcalcifications, were retrospectively collected. Immunohistochemical analysis was performed to evaluate the expression of vimentin, BMP-2, BMP-4, RANKL, Runx2, OPN, PTX3, and SDF-1, while Kaplan-Meier plots were used to assess their prognostic impact on overall survival in a dataset of 2976 breast cancer patients. The expression of vimentin, BMP-2, BMP-4, and SDF-1 was significantly higher in malignant lesions compared to benign ones, regardless of the presence of microcalcifications. Notably, these markers showed no correlation with traditional prognostic factors, such as tumor grade or hormone receptor status. The bioinformatics analysis provided valuable insights into the possible prognostic and therapeutic significance of BMP-2, BMP-4, SDF-1, and vimentin in breast cancer. In fact, all these biomarkers impact on the overall survival in specific molecular breast cancer types. In addition, high expression of SDF-1 and vimentin is able to predict the response to chemotherapy. The findings here reported suggest that vimentin, BMP-2, BMP-4, and SDF-1 could be independent prognostic biomarkers in breast cancer, providing insights beyond traditional clinical factors.
Collapse
Affiliation(s)
- Erica Giacobbi
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Rita Bonfiglio
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Gabriele Rotondaro
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Francesca Servadei
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Valeria Palumbo
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Maria Paola Scioli
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Elena Bonanno
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Claudio Oreste Buonomo
- Breast Unit, Department of Surgical Science, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.O.B.); (G.V.)
| | - Gianluca Vanni
- Breast Unit, Department of Surgical Science, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.O.B.); (G.V.)
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| | - Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (E.G.); (R.B.); (G.R.); (F.S.); (A.S.); (V.P.); (M.P.S.); (E.B.); (E.C.); (M.S.)
| |
Collapse
|
4
|
Zhao G, Zhao X, Liu Z, Wang B, Dong P, Watari H, Pfeffer LM, Tigyi G, Zhang W, Yue J. Knockout or inhibition of DHPS suppresses ovarian tumor growth and metastasis by attenuating the TGFβ pathway. Sci Rep 2025; 15:917. [PMID: 39762448 PMCID: PMC11704301 DOI: 10.1038/s41598-025-85466-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Deoxyhypusine synthase (DHPS) is an enzyme encoded by the DHPS gene, with high expression in various cancers, including ovarian cancer (OC). DHPS regulates the translation initiation factor EIF5A, and EIF5A2 knockout inhibits OC tumor growth and metastasis by blocking the epithelial-to-mesenchymal transition (EMT) and the TGFβ pathway. In this study, we show that DHPS is amplified in OC patients, and its elevated expression correlates with poor survival. Using lentiviral CRISPR/Cas9 vectors for DHPS knockout, we observed EMT inhibition in SKOV3 and OVCAR8 cells through suppressed hypusination and reduced EIF5A2 expression. Inhibition of DHPS activity with GC7 similarly blocked hypusination and EMT. Disrupting DHPS expression, either genetically or pharmacologically, inhibited primary tumor growth and metastasis in OC mouse models. These findings suggest that targeting DHPS and inhibiting hypusination could be promising strategies for OC treatment.
Collapse
Affiliation(s)
- Guannan Zhao
- Department of Pathology and Laboratory Medicine, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, United States
- Center for Cancer Research, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Xinxin Zhao
- Department of Obstetrics and Gynecology, The Third Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Ziping Liu
- Department of Obstetrics and Gynecology, The Third Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Baojin Wang
- Department of Obstetrics and Gynecology, The Third Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, United States
- Center for Cancer Research, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Gabor Tigyi
- Department of Physiology, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Wenjing Zhang
- Department of Genetics, Genomics & Informatics, Collage of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, United States.
- Center for Cancer Research, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
5
|
Wu Y, Rong L, Zhang S, He Y, Song N, Zuo G, Mei Z. Ligustilide Inhibits the PI3K/AKT Signalling Pathway and Suppresses Cholangiocarcinoma Cell Proliferation, Migration, and Invasion. Recent Pat Anticancer Drug Discov 2025; 20:200-212. [PMID: 39171465 DOI: 10.2174/0115748928332384240812060751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Angelica sinensis (Oliv.) Diels, a renowned traditional Chinese medicine, has gained widespread recognition for its antitumor properties. Further investigation is warranted to determine whether ligustilide (LIG), which is extracted from this plant, can effectively inhibit tumors. OBJECTIVES We delved into the impact of LIG on cholangiocarcinoma cells, aiming to unravel the mechanisms underlying its effects. MATERIALS AND METHODS Cholangiocarcinoma cells (HuccT1 and RBE) were exposed to varying concentrations of LIG (2, 5, 10, 15, 20 μg/mL) for 24, 48, and 72 h. After identifying differentially expressed genes, stable transcription strains were utilized to explore LIG's antitumor mechanism. The inhibitory effects of LIG (5 μg/mL, 48 h) were assessed by CCK-8, colony formation, wound healing, transwell migration, western blotting, and immunofluorescence. In vivo, experiments in NOG mice (Ac, Ac+LIG; five per group) evaluated LIG's antiproliferative efficacy (5 mg/kg, intraperitoneal injection, 18-day period). RESULTS LIG significantly inhibited cell proliferation and migration with IC50 5.08 and 5.77 μg/mL in HuccT1 and RBE cell lines at 48h, increased the expression of E-cadherin while decreased N-cadherin and the protein of PI3K/AKT pathway. Silenced NDRG1 (N-Myc downstream- regulated gene 1) attenuated these effects. In vivo, the AC+LIG group (LIG, 5 mg/kg, qd, 18 d) exhibited smaller tumor volumes compared to the Ac group. The expression of Ki-67 was significantly downregulated in the AC+LIG group. CONCLUSION For the first time, our study has revealed that LIG holds therapeutic potential for treating cholangiocarcinoma. These findings hold promise for advancing innovative therapeutic approaches in the treatment of cholangiocarcinoma. LIG may serve as a useful patent for treating CCA.
Collapse
Affiliation(s)
- Yue Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Rong
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Suifeng Zhang
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Yuxi He
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Na Song
- Department of Oncology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Guoqing Zuo
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhechuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Yuan J, Yang L, Zhang H, Beeraka NM, Zhang D, Wang Q, Wang M, Pr HV, Sethi G, Wang G. Decoding tumor microenvironment: EMT modulation in breast cancer metastasis and therapeutic resistance, and implications of novel immune checkpoint blockers. Biomed Pharmacother 2024; 181:117714. [PMID: 39615165 DOI: 10.1016/j.biopha.2024.117714] [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: 08/14/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Tumor microenvironment (TME) and epithelial-mesenchymal transition (EMT) play crucial roles in the initiation and progression of tumors. TME is composed of various cell types, such as immune cells, fibroblasts, and endothelial cells, as well as non-cellular components like extracellular matrix (ECM) proteins and soluble factors. These elements interact with tumor cells through a complex network of signaling pathways involving cytokines, growth factors, metabolites, and non-coding RNA-carrying exosomes. Hypoxic conditions within the TME further modulate these interactions, collectively influencing tumor growth, metastatic potential, and response to therapy. EMT represents a dynamic and reversible process where epithelial cells undergo phenotypic changes to adopt mesenchymal characteristics in several cancers, including breast cancers. This transformation enhances cell motility and imparts stem cell-like properties, which are closely associated with increased metastatic capability and resistance to conventional cancer treatments. Thus, understanding the crosstalk between the TME and EMT is essential for unraveling the underlying mechanisms of breast cancer metastasis and therapeutic resistance. This review uniquely examines the intricate interplay between the tumor TME and epithelial-mesenchymal transition EMT in driving breast cancer metastasis and treatment resistance. It explores the therapeutic potential of targeting the TME-EMT axis, specifically through CD73-TGF-β dual-blockade, to improve outcomes in triple-negative breast cancer. Additionally, it underscores new strategies to enhance immune checkpoint blockade (ICB) responses by modulating EMT, thereby offering innovative insights for more effective cancer treatment.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Li Yang
- Department of Clinical Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Hua Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Narasimha M Beeraka
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia; Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu, Anantapuramu, Andhra Pradesh 515721, India; Department of Studies in Molecular Biology, Faculty of Science and Technology, University of Mysore, Mysore, Karnataka, 570006, India.
| | - Danfeng Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Qun Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Minghua Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Hemanth Vikram Pr
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Geng Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| |
Collapse
|
7
|
Kuo LW, Crump LS, O’Neill K, Williams MM, Christenson JL, Spoelstra NS, Roy MK, Argabright A, Reisz JA, D’Alessandro A, Boorgula MP, Goodspeed A, Bickerdike M, Bitler BG, Richer JK. Blocking Tryptophan Catabolism Reduces Triple-Negative Breast Cancer Invasive Capacity. CANCER RESEARCH COMMUNICATIONS 2024; 4:2699-2713. [PMID: 39311710 PMCID: PMC11484926 DOI: 10.1158/2767-9764.crc-24-0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/26/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
SIGNIFICANCE TDO2 is more highly expressed than the nonhomologous TRP-catabolizing enzyme IDO1 in TNBC. We find that TDO2 knockdown can lead to a compensatory increase in IDO1. Therefore, we tested a newly developed TDO2/IDO1 dual inhibitor and found that it decreases TRP catabolism, anchorage-independent survival, and invasive capacity.
Collapse
Affiliation(s)
- Li-Wei Kuo
- Cancer Biology Training Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Lyndsey S. Crump
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Kathleen O’Neill
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Michelle M. Williams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Jessica L. Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Nicole S. Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Micaela Kalani Roy
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Amy Argabright
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Meher P. Boorgula
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Andrew Goodspeed
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | | | - Benjamin G. Bitler
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Jennifer K. Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
8
|
García-Hernández AP, Sánchez-Sánchez G, Carlos-Reyes A, López-Camarillo C. Functional roles of microRNAs in vasculogenic mimicry and resistance to therapy in human cancers: an update. Expert Rev Clin Immunol 2024; 20:913-926. [PMID: 38712535 DOI: 10.1080/1744666x.2024.2352484] [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: 12/06/2023] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
INTRODUCTION Vasculogenic mimicry (VM) alludes to the ability of cancer cells to organize on three-dimensional channel-like structures to obtain nutrients and oxygen. This mechanism confers an aggressive phenotype, metastatic potential, and resistance to chemotherapy resulting in a poor prognosis. Recent studies have been focused on the identification of microRNAs (miRNAs) that regulate the VM representing potential therapeutic targets in cancer. AREAS COVERED An overview of the roles of miRNAs on VM development and their functional relationships with tumor microenvironment. The functions of cancer stem-like cells in VM, and resistance to therapy are also discussed. Moreover, the modulation of VM by natural compounds is explored. The clinical significance of deregulated miRNAs as potential therapeutic targets in tumors showing VM is further highlighted. EXPERT OPINION The miRNAs are regulators of protein-encoding genes involved in VM; however, their specific expression signatures with clinical value in large cohorts of patients have not been established yet. We considered that genomic profiling of miRNAs could be useful to define some hallmarks of tumors such as stemness, drug resistance, and VM in cancer patients. However, additional studies are needed to establish the relevant role of miRNAs as effective therapeutic targets in tumors that have developed VM.
Collapse
Affiliation(s)
| | | | - Angeles Carlos-Reyes
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Ciudad de México
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México
| |
Collapse
|
9
|
Coelho LL, Vianna MM, da Silva DM, Gonzaga BMDS, Ferreira RR, Monteiro AC, Bonomo AC, Manso PPDA, de Carvalho MA, Vargas FR, Garzoni LR. Spheroid Model of Mammary Tumor Cells: Epithelial-Mesenchymal Transition and Doxorubicin Response. BIOLOGY 2024; 13:463. [PMID: 39056658 PMCID: PMC11273983 DOI: 10.3390/biology13070463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 07/28/2024]
Abstract
Breast cancer is the most prevalent cancer among women worldwide. Therapeutic strategies to control tumors and metastasis are still challenging. Three-dimensional (3D) spheroid-type systems more accurately replicate the features of tumors in vivo, working as a better platform for performing therapeutic response analysis. This work aimed to characterize the epithelial-mesenchymal transition and doxorubicin (dox) response in a mammary tumor spheroid (MTS) model. We evaluated the doxorubicin treatment effect on MCF-7 spheroid diameter, cell viability, death, migration and proteins involved in the epithelial-mesenchymal transition (EMT) process. Spheroids were also produced from tumors formed from 4T1 and 67NR cell lines. MTSs mimicked avascular tumor characteristics, exhibited adherens junction proteins and independently produced their own extracellular matrix. Our spheroid model supports the 3D culturing of cells isolated from mice mammary tumors. Through the migration assay, we verified a reduction in E-cadherin expression and an increase in vimentin expression as the cells became more distant from spheroids. Dox promoted cytotoxicity in MTSs and inhibited cell migration and the EMT process. These results suggest, for the first time, that this model reproduces aspects of the EMT process and describes the potential of dox in inhibiting the metastatic process, which can be further explored.
Collapse
Affiliation(s)
- Laura Lacerda Coelho
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Matheus Menezes Vianna
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Debora Moraes da Silva
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Beatriz Matheus de Souza Gonzaga
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Roberto Rodrigues Ferreira
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Ana Carolina Monteiro
- Laboratory of Osteo and Tumor Immunology, Department of Immunobiology, Fluminense Federal University (UFF), Rio de Janeiro 24020-150, Brazil;
- Thymus Research Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Adriana Cesar Bonomo
- Thymus Research Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Pedro Paulo de Abreu Manso
- Laboratory of Pathology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | | | - Fernando Regla Vargas
- Laboratory of Epidemiology of Congenital Malformations, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Luciana Ribeiro Garzoni
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| |
Collapse
|
10
|
Luo L, Xu N, Fan W, Wu Y, Chen P, Li Z, He Z, Liu H, Lin Y, Zheng G. The TGFβ2-Snail1-miRNA TGFβ2 Circuitry is Critical for the Development of Aggressive Functions in Breast Cancer. Clin Transl Med 2024; 14:e1558. [PMID: 38299307 PMCID: PMC10831563 DOI: 10.1002/ctm2.1558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
There have been contradictory reports on the biological role of transforming growth factor-βs (TGFβs) in breast cancer (BC), especially with regard to their ability to promote epithelial-mesenchymal transition (EMT). Here, we show that TGFβ2 is preferentially expressed in mesenchymal-like BCs and maintains the EMT phenotype, correlating with cancer stem cell-like characteristics, growth, metastasis and chemo-resistance and predicting worse clinical outcomes. However, this is only true in ERα- BC. In ERα+ luminal-type BC, estrogen receptor interacts with p-Smads to block TGFβ signalling. Furthermore, we also identify a microRNAs (miRNAs) signature (miRNAsTGFβ2 ) that is weakened in TGFβ2-overexpressing BC cells. We discover that TGFβ2-Snail1 recruits enhancer of zeste homolog-2 to convert miRNAsTGFβ2 promoters from an active to repressive chromatin configuration and then repress miRNAsTGFβ2 transcription, forming a negative feedback loop. On the other hand, miRNAsTGFβ2 overexpression reverses the mesenchymal-like traits in agreement with the inhibition of TGFβ2-Snail1 signalling in BC cells. These findings clarify the roles of TGFβ2 in BC and suggest novel therapeutic strategies based on the TGFβ2-Snail1-miRNAsTGFβ2 loop for a subset type of human BCs.
Collapse
Affiliation(s)
- Liyun Luo
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Ning Xu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Weina Fan
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Yixuan Wu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Pingping Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Zhihui Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Zhimin He
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Hao Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Ying Lin
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Guopei Zheng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| |
Collapse
|
11
|
Vydra N, Toma-Jonik A, Janus P, Mrowiec K, Stokowy T, Głowala-Kosińska M, Sojka DR, Olbryt M, Widłak W. An Increase in HSF1 Expression Directs Human Mammary Epithelial Cells toward a Mesenchymal Phenotype. Cancers (Basel) 2023; 15:4965. [PMID: 37894333 PMCID: PMC10605143 DOI: 10.3390/cancers15204965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
HSF1 is a well-known heat shock protein expression regulator in response to stress. It also regulates processes important for growth, development or tumorigenesis. We studied the HSF1 influence on the phenotype of non-tumorigenic human mammary epithelial (MCF10A and MCF12A) and several triple-negative breast cancer cell lines. MCF10A and MCF12A differ in terms of HSF1 levels, morphology, growth in Matrigel, expression of epithelial (CDH1) and mesenchymal (VIM) markers (MCF10A are epithelial cells; MCF12A resemble mesenchymal cells). HSF1 down-regulation led to a reduced proliferation rate and spheroid formation in Matrigel by MCF10A cells. However, it did not affect MCF12A proliferation but led to CDH1 up-regulation and the formation of better organized spheroids. HSF1 overexpression in MCF10A resulted in reduced CDH1 and increased VIM expression and the acquisition of elongated fibroblast-like morphology. The above-mentioned results suggest that elevated levels of HSF1 may direct mammary epithelial cells toward a mesenchymal phenotype, while a lowering of HSF1 could reverse the mesenchymal phenotype to an epithelial one. Therefore, HSF1 may be involved in the remodeling of mammary gland architecture over the female lifetime. Moreover, HSF1 levels positively correlated with the invasive phenotype of triple-negative breast cancer cells, and their growth was inhibited by the HSF1 inhibitor DTHIB.
Collapse
Affiliation(s)
- Natalia Vydra
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-102 Gliwice, Poland; (A.T.-J.); (P.J.); (K.M.); (M.G.-K.); (D.R.S.); (M.O.)
| | - Agnieszka Toma-Jonik
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-102 Gliwice, Poland; (A.T.-J.); (P.J.); (K.M.); (M.G.-K.); (D.R.S.); (M.O.)
| | - Patryk Janus
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-102 Gliwice, Poland; (A.T.-J.); (P.J.); (K.M.); (M.G.-K.); (D.R.S.); (M.O.)
| | - Katarzyna Mrowiec
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-102 Gliwice, Poland; (A.T.-J.); (P.J.); (K.M.); (M.G.-K.); (D.R.S.); (M.O.)
| | - Tomasz Stokowy
- Scientific Computing Group, IT Division, University of Bergen, N-5008 Bergen, Norway;
| | - Magdalena Głowala-Kosińska
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-102 Gliwice, Poland; (A.T.-J.); (P.J.); (K.M.); (M.G.-K.); (D.R.S.); (M.O.)
| | - Damian Robert Sojka
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-102 Gliwice, Poland; (A.T.-J.); (P.J.); (K.M.); (M.G.-K.); (D.R.S.); (M.O.)
| | - Magdalena Olbryt
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-102 Gliwice, Poland; (A.T.-J.); (P.J.); (K.M.); (M.G.-K.); (D.R.S.); (M.O.)
| | - Wiesława Widłak
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-102 Gliwice, Poland; (A.T.-J.); (P.J.); (K.M.); (M.G.-K.); (D.R.S.); (M.O.)
| |
Collapse
|
12
|
Eskandari N, Gentile S. Potassium channels activity unveils cancer vulnerability. CURRENT TOPICS IN MEMBRANES 2023; 92:1-14. [PMID: 38007264 DOI: 10.1016/bs.ctm.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
"No cell could exist without ion channels" (Clay Armstrong; 1999). Since the discovery in the early 1950s, that ions move across biological membranes, the idea that changes of ionic gradients can generate biological signals has fascinated scientists in any fields. Soon later (1960s) it was found that ionic flows were controlled by a class of specific and selective proteins called ion channels. Thus, it became clear that the concerted activities of these proteins can initiate, arrest, and finely tune a variety of biochemical cascades which offered the opportunity to better understand both biology and pathology. Cancer is a disease that is notoriously difficult to treat due its heterogeneous nature which makes it the deadliest disease in the developed world. Recently, emerging evidence has established that potassium channels are critical modulators of several hallmarks of cancer including tumor growth, metastasis, and metabolism. Nevertheless, the role of potassium ion channels in cancer biology and the therapeutic potential offered by targeting these proteins has not been explored thoroughly. This chapter is addressed to both cancer biologists and ion channels scientists and it aims to shine a light on the established and potential roles of potassium ion channels in cancer biology and on the therapeutic benefit of targeting potassium channels with activator molecules.
Collapse
Affiliation(s)
- Najmeh Eskandari
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Saverio Gentile
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
13
|
Shiino S, Tokura M, Nakayama J, Yoshida M, Suto A, Yamamoto Y. Investigation of Tumor Heterogeneity Using Integrated Single-Cell RNA Sequence Analysis to Focus on Genes Related to Breast Cancer-, EMT-, CSC-, and Metastasis-Related Markers in Patients with HER2-Positive Breast Cancer. Cells 2023; 12:2286. [PMID: 37759508 PMCID: PMC10527746 DOI: 10.3390/cells12182286] [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: 08/23/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) protein, which is characterized by the amplification of ERBB2, is a molecular target for HER2-overexpressing breast cancer. Many targeted HER2 strategies have been well developed thus far. Furthermore, intratumoral heterogeneity in HER2 cases has been observed with immunohistochemical staining and has been considered one of the reasons for drug resistance. Therefore, we conducted an integrated analysis of the breast cancer single-cell gene expression data for HER2-positive breast cancer cases from both scRNA-seq data from public datasets and data from our cohort and compared them with those for luminal breast cancer datasets. In our results, heterogeneous distribution of the expression of breast cancer-related genes (ESR1, PGR, ERBB2, and MKI67) was observed. Various gene expression levels differed at the single-cell level between the ERBB2-high group and ERBB2-low group. Moreover, molecular functions and ERBB2 expression levels differed between estrogen receptor (ER)-positive and ER-negative HER2 cases. Additionally, the gene expression levels of typical breast cancer-, CSC-, EMT-, and metastasis-related markers were also different across each patient. These results suggest that diversity in gene expression could occur not only in the presence of ERBB2 expression and ER status but also in the molecular characteristics of each patient.
Collapse
Affiliation(s)
- Sho Shiino
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Momoko Tokura
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan; (M.T.); (J.N.)
| | - Jun Nakayama
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan; (M.T.); (J.N.)
| | - Masayuki Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Akihiko Suto
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Yusuke Yamamoto
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan; (M.T.); (J.N.)
| |
Collapse
|
14
|
Feng Y, McGuire N, Walton A, AP-MBC Consortium, Fox S, Papa A, Lakhani SR, McCart Reed AE. Predicting breast cancer-specific survival in metaplastic breast cancer patients using machine learning algorithms. J Pathol Inform 2023; 14:100329. [PMID: 37664452 PMCID: PMC10470383 DOI: 10.1016/j.jpi.2023.100329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Metaplastic breast cancer (MpBC) is a rare and aggressive subtype of breast cancer, with data emerging on prognostic factors and survival prediction. This study aimed to develop machine learning models to predict breast cancer-specific survival (BCSS) in MpBC patients, utilizing a dataset of 160 patients with clinical, pathological, and biological variables. An in-depth variable selection process was carried out using gain ratio and correlation-based methods, resulting in 10 variables for model estimation. Five models (decision tree with bagging; logistic regression; multilayer perceptron; naïve Bayes; and, random forest algorithms) were evaluated using 10-fold cross-validation. Despite the constraints posed by the absence of therapeutic information, the random forest model exhibited the highest performance in predicting BCSS, with an ROC area of 0.808. This study emphasizes the potential of machine learning algorithms in predicting prognosis for complex and heterogeneous cancer subtypes using clinical datasets, and their potential to contribute to patient management. Further research that incorporates additional variables, such as treatment response, and more advanced machine learning techniques will likely enhance the predictive power of MpBC prognostic models.
Collapse
Affiliation(s)
- Yufan Feng
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane 4029, Australia
| | - Natasha McGuire
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane 4029, Australia
| | - Alexandra Walton
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane 4029, Australia
- Pathology Queensland, The Royal Brisbane and Women’s Hospital, Brisbane 4029, Australia
| | | | - Stephen Fox
- Peter MacCallum Cancer Centre and University of Melbourne, Melbourne 3000, Australia
| | - Antonella Papa
- Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Sunil R. Lakhani
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane 4029, Australia
- Pathology Queensland, The Royal Brisbane and Women’s Hospital, Brisbane 4029, Australia
| | - Amy E. McCart Reed
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane 4029, Australia
| |
Collapse
|
15
|
Li S, He J, Liao X, He Y, Chen R, Chen J, Hu S, Sun J. Fbxo22 inhibits metastasis in triple-negative breast cancer through ubiquitin modification of KDM5A and regulation of H3K4me3 demethylation. Cell Biol Toxicol 2023; 39:1641-1655. [PMID: 36112263 PMCID: PMC10425479 DOI: 10.1007/s10565-022-09754-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022]
Abstract
The importance of Fbxo22 in carcinogenesis has been highly documented. Here, we discussed downstream regulatory factors of Fbxo22 in TNBC. RNA-sequencing was conducted for identifying differentially expressed genes, followed by construction of a regulatory network. Expression patterns of Fbxo22/KDM5A in TNBC were determined by their correlation with the prognosis analyzed. Then, regulation mechanisms between Fbxo22 and KDM5A as well as between KDM5A and H3K4me3 were assayed. After silencing and overexpression experiments, the significance of Fbxo22 in repressing tumorigenesis in vitro and in vivo was explored. Fbxo22 was poorly expressed, while KDM5A was highly expressed in TNBC. Patients with elevated Fbxo22, decreased KDM5A, or higher p16 had long overall survival. Fbxo22 reduced the levels of KDM5A by ubiquitination. KDM5A promoted histone H3K4me3 demethylation to downregulate p16 expression. Fbxo22 reduced KDM5A expression to enhance p16, thus inducing DNA damage as well as reducing tumorigenesis and metastasis in TNBC. Our study validated FBXO22 as a tumor suppressor in TNBC through ubiquitination of KDM5A and regulation of p16.
Collapse
Affiliation(s)
- Siqiaozhi Li
- Shenzhen Toyon Biotechnology Co., Ltd, Shenzhen, 518057, People's Republic of China
- Shenzhen Beike Biotechnology Research Institute, Shenzhen, 518057, People's Republic of China
| | - Jinsong He
- Department of Breast Surgery, Shenzhen Hospital of Peking University, Shenzhen, 518057, People's Republic of China
| | - Xin Liao
- Shenzhen Toyon Biotechnology Co., Ltd, Shenzhen, 518057, People's Republic of China
| | - Yixuan He
- Shenzhen Toyon Biotechnology Co., Ltd, Shenzhen, 518057, People's Republic of China
| | - Rui Chen
- Shenzhen Toyon Biotechnology Co., Ltd, Shenzhen, 518057, People's Republic of China
| | - Junhui Chen
- Intervention and Cell Therapy Center, Shenzhen Hospital of Peking University, No. 1120, Lianhua Road, Shenzhen, 518057, Guangdong Province, People's Republic of China
| | - Sean Hu
- Shenzhen Beike Biotechnology Research Institute, Shenzhen, 518057, People's Republic of China
| | - Jia Sun
- Shenzhen Toyon Biotechnology Co., Ltd, Shenzhen, 518057, People's Republic of China.
- Intervention and Cell Therapy Center, Shenzhen Hospital of Peking University, No. 1120, Lianhua Road, Shenzhen, 518057, Guangdong Province, People's Republic of China.
| |
Collapse
|
16
|
Fang Q, Xin W, Chen L, Fu Y, Qi Y, Ding H, Fang L. Caffeic acid phenethyl ester suppresses metastasis of breast cancer cells by inactivating FGFR1 via MD2. PLoS One 2023; 18:e0289031. [PMID: 37490511 PMCID: PMC10368285 DOI: 10.1371/journal.pone.0289031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Tumor metastasis is the main cause of death for breast cancer patients. Caffeic acid phenethyl ester (CAPE) has strong anti-tumor effects with very low toxicity and may be a potential candidate drug. However, the anti-metastatic effect and molecular mechanism of CAPE on breast cancer need more research. METHODS MCF-7 and MDA-MB-231 breast cancer cells were used here. Wound healing and Transwell assay were used for migration and invasion detection. Western blot and RT-qPCR were carried out for the epithelial-to-myofibroblast transformation (EMT) process investigation. Western blot and immunofluorescence were performed for fibroblast growth factor receptor1 (FGFR1) phosphorylation and nuclear transfer detection. Co-immunoprecipitation was used for the FGFR1/myeloid differentiation protein2 (MD2) complex investigation. RESULTS Our results suggested that CAPE blocks the migration, invasion, and EMT process of breast cancer cells. Mechanistically, CAPE inhibits FGFR1 phosphorylation and nuclear transfer while overexpression of FGFR1 reduces the anti-metastasis effect of CAPE. Further, we found that FGFR1 is bound to MD2, and silencing MD2 inhibits FGFR1 phosphorylation and nuclear transfer as well as cell migration and invasion. CONCLUSION This study illustrated that CAPE restrained FGFR1 activation and nuclear transfer through MD2/FGFR1 complex inhibition and showed good inhibitory effects on the metastasis of breast cancer cells.
Collapse
Affiliation(s)
- Qilu Fang
- Zhejiang Cancer Hospital, Hangzhou Institute Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wenxiu Xin
- Zhejiang Cancer Hospital, Hangzhou Institute Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base of Zhejiang Cancer Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liangsheng Chen
- Postgraduate Training Base of Zhejiang Cancer Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxuan Fu
- Postgraduate Training Base of Zhejiang Cancer Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yajun Qi
- Zhejiang Cancer Hospital, Hangzhou Institute Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Haiying Ding
- Zhejiang Cancer Hospital, Hangzhou Institute Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Luo Fang
- Zhejiang Cancer Hospital, Hangzhou Institute Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base of Zhejiang Cancer Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
17
|
Sun R, He J, Xiang Q, Feng Y, Gong Y, Ning Y, Deng C, Sun K, Zhang M, Cheng Z, Le X, Xiong Q, Dai F, Wu Y, Xiang T. NTF4 plays a dual role in breast cancer in mammary tumorigenesis and metastatic progression. Int J Biol Sci 2023; 19:641-657. [PMID: 36632451 PMCID: PMC9830504 DOI: 10.7150/ijbs.79435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023] Open
Abstract
Breast cancer metastasis can happen even when the primary tumor is relatively small. But the mechanism for such early metastasis is poorly understood. Herein, we report that neurotrophin 4 (NTF4) plays a dual role in breast cancer proliferation and metastasis. Clinical data showed high levels of NTF4, especially in the early stage, to be associated with poor clinical outcomes, supporting the notion that metastasis, rather than primary cancer, was the major determinant of breast cancer mortality for patients. NTF4 promoted epithelial-mesenchymal transition (EMT), cell motility, and invasiveness of breast cancer cells in vitro and in vivo. Interestingly, NTF4 inhibited cell proliferation while promoting cellular apoptosis in vitro and inhibited xenograft tumorigenicity in vivo. Mechanistically, NTF4 elicited its pro-metastatic effects by activating PRKDC/AKT and ANXA1/NF-κB pathways to stabilize SNAIL protein, therefore decreasing the level of E-cadherin. Conversely, NTF4 increased ANXA1 phosphorylation and sumoylation and the interaction with importin β, leading to nuclear import and retention of ANXA1, which in turn activates the caspase-3 apoptosis cascade. Our findings identified an unexpected dual role for NTF4 in breast cancer which contributes to early metastasis of the disease. Therefore, NTF4 may serve as a prognostic marker and a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Ran Sun
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.,Department of Oncology, Jiulongpo People's Hospital, Chongqing 400050, China
| | - Jin He
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qin Xiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yixiao Feng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yijia Gong
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yijiao Ning
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chaoqun Deng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Kexin Sun
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mingjun Zhang
- Department of Laboratory Medicine, Jiulongpo People's Hospital, Chongqing 400050, China
| | - Zhaobo Cheng
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xin Le
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qi Xiong
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fengsheng Dai
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yongzhong Wu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China.,✉ Corresponding authors: Tingxiu Xiang. Tel: (023) 65079282. E-mail: and Yongzhong Wu. E-mail:
| | - Tingxiu Xiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.,Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China.,✉ Corresponding authors: Tingxiu Xiang. Tel: (023) 65079282. E-mail: and Yongzhong Wu. E-mail:
| |
Collapse
|
18
|
Kim K, Lee J, Lee JY, Yong SH, Kim EY, Jung JY, Kang YA, Park MS, Kim YS, Oh CM, Lee SH. Clinical features and molecular genetics associated with brain metastasis in suspected early-stage non-small cell lung cancer. Front Oncol 2023; 13:1148475. [PMID: 37139160 PMCID: PMC10150586 DOI: 10.3389/fonc.2023.1148475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Regarding whether brain magnetic resonance imaging (MRI) should be routine in patients with suspected early-stage lung cancer, guideline recommendations are inconsistent. Therefore, we performed this study to evaluate the incidence of and risk factors for brain metastasis (BM) in patients with suspected early-stage non-small-cell lung cancer (NSCLC). Methods A review of the medical charts of consecutive NSCLC patients diagnosed between January 2006 and May 2020 was performed. We identified 1,382 NSCLC patients with clinical staging of T1/2aN0M0 (excluding BM), and investigated the incidence, clinical predictors, and prognosis of BM in the cohort. We also performed RNA-sequencing differential expression analysis using transcriptome of 8 patients, using DESeq2 package (version 1.32.0) with R (version 4.1.0). Results Among 1,382 patients, nine hundred forty-nine patients (68.7%) underwent brain MRI during staging, and 34 patients (3.6%) were shown to have BM. Firth's bias-reduced logistic regression showed that tumor size (OR 1.056; 95% CI 1.009-1.106, p=0.018) was the only predictor of BM, and pathologic type was not a predictor of BM in our cohort (p>0.05). The median overall survival for patients with brain metastasis was 5.5 years, which is better than previously reported in the literature. RNA-sequencing differential expression analysis revealed the top 10 significantly upregulated genes and top 10 significantly downregulated genes. Among the genes involved in BM, Unc-79 homolog, non-selective sodium leak channel (NALCN) channel complex subunit (UNC79) was the most highly expressed gene in the lung adenocarcinoma tissues from the BM group, and an in vitro assay using A549 cells revealed that the NALCN inhibitor suppressed lung cancer cell proliferation and migration. Conclusions Given the incidence and favorable outcome of BM in patients with suspected early-stage NSCLC, selective screening with brain MRI may be considered, especially in patients with high-risk features.
Collapse
Affiliation(s)
- Kangjoon Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jibeom Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jeong-Yun Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Seung Hyun Yong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Ye Jung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Ae Kang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moo Suk Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Sam Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- *Correspondence: Chang-Myung Oh, ; Sang Hoon Lee,
| | - Sang Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Chang-Myung Oh, ; Sang Hoon Lee,
| |
Collapse
|
19
|
EMT Molecular Signatures of Pancreatic Neuroendocrine Neoplasms. Int J Mol Sci 2022; 23:ijms232113645. [PMID: 36362433 PMCID: PMC9657865 DOI: 10.3390/ijms232113645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Neuroendocrine neoplasms (NENs) are relatively rare neoplasms occurring predominantly in the gastrointestinal tract and pancreas. Their heterogeneity poses challenges for diagnosis and treatment. There is a paucity of markers for characterisation of NEN tumours. For routine diagnosis, immunohistochemistry of the NEN-specific markers CgA and synaptophysin and the proliferation marker Ki-67 are used. These parameters, however, are qualitative and lack the capacity to fully define the tumour phenotype. Molecules of epithelial–mesenchymal transition (EMT) are potential candidates for improved tumour characterisation. Using qRT-PCR, we measured mRNA levels of 27 tumour markers, including 25 EMT-associated markers, in tumour tissue and matched non-tumour tissues for 13 patients with pancreatic NENs. Tissue from patients with three different grades of tumour had distinctly different mRNA profiles. Of the 25 EMT-associated markers analysed, 17 were higher in G3 tissue relative to matched non-tumour tissue, including CD14, CD24, CD31, CD44, CD45, CD56, CK6, CK7, CK13, CK20, NSE, CDX2, CgA, DAXX, PCNA, laminin and Ki-67. The differences in levels of seven EMT-associated markers, Ki-67, DAXX, CD24, CD44, vimentin, laminin and PDX1 plus CgA and NSE (neuroendocrine markers) enabled a distinct molecular signature for each tumour grade to be generated. EMT molecules differentially expressed in three tumour grades have potential for use in tumour stratification and prognostication and as therapeutic targets for treatment of neuroendocrine cancers, following validation with additional samples.
Collapse
|
20
|
Clonal evolution and expansion associated with therapy resistance and relapse of colorectal cancer. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 790:108445. [PMID: 36371022 DOI: 10.1016/j.mrrev.2022.108445] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Colorectal cancer (CRC) arises by a continuous process of genetic diversification and clonal evolution. Multiple genes and pathways have a role in tumor initiation and progression. The gradual accumulation of genetic and epigenetic processes leads to the establishment of adenoma and cancer. The important 'driver' mutations in tumor suppressor genes (such as TP53, APC, and SMAD4) and oncogenes (such as KRAS, NRAS, MET, and PIK3CA) confer selective growth advantages and cause CRC advancement. Clonal evolution induced by therapeutic pressure, as well as intra-tumoral heterogeneity, has been a great challenge in the treatment of metastatic CRC. Tumors often develop resistance to treatments as a result of intra-tumor heterogeneity, clonal evolution, and selection. Hence, the development of a multidrug personalized approach should be prioritized to pave the way for therapeutics repurposing and combination therapy to arrest tumor progression. This review summarizes how selective drug pressure can impact tumor evolution, resulting in the formation of polyclonal resistance mechanisms, ultimately promoting cancer progression. Current strategies for targeting clonal evolution are described. By understanding sources and consequences of tumor heterogeneity, customized and effective treatment plans to combat drug resistance may be devised.
Collapse
|
21
|
Dong J, Pervaiz W, Tayyab B, Li D, Kang L, Zhang H, Gong H, Ma X, Li J, Agboyibor C, Bi Y, Liu H. A comprehensive comparative study on LSD1 in different cancers and tumor specific LSD1 inhibitors. Eur J Med Chem 2022; 240:114564. [PMID: 35820351 DOI: 10.1016/j.ejmech.2022.114564] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 01/14/2023]
Abstract
LSD1 was significantly over-expressed in several cancer types, and its aberrant overexpression was revealed to play a crucial role in the initiation and progression of cancer. Several LSD1 inhibitors that were discovered and developed so far were found to be effective in attenuating tumor growth in both in vivo and in vitro studies. However, the major challenge associated with the development of cancer therapies is personalized treatment. Therefore, it is essential to look in detail at how LSD1 plays its part in carcinogenesis and whether there are any different expression levels of LSD1 in different tumors. Here in this review, fresh insight into a list of function correlated LSD1 binding proteins are provided, and we tried to figure out the role of LSD1 in different cancer types, including hematological malignancies and solid tumors. A critical description of mutation preference for LSD1 in different tumors was also discussed. Recent research findings clearly showed that the abrogation of LSD1 demethylase activity via LSD1 inhibitors markedly reduced the growth of cancer cells. But there are still many ambiguities regarding the role of LSD1 in different cancers. Therefore, targeting LSD1 for treating different cancers is still reductionist, and many challenges need to be met to improve the therapeutic outcomes of LSD1 inhibitors.
Collapse
Affiliation(s)
- Jianshu Dong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China.
| | - Waqar Pervaiz
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Bilal Tayyab
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Dié Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Lei Kang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Huimin Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Huimin Gong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Xinli Ma
- China-US(Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan, 450008, China
| | - Jian Li
- China-US(Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan, 450008, China
| | - Clement Agboyibor
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuefeng Bi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China.
| | - Hongmin Liu
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
22
|
Hager E, Chen J, Zhao L. Minireview: Parabens Exposure and Breast Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:1873. [PMID: 35162895 PMCID: PMC8834979 DOI: 10.3390/ijerph19031873] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/21/2022]
Abstract
There is increasing recognition that environmental exposure to chemicals, such as endocrine-disruptive chemicals (EDCs), contributes to the development of breast cancer. Parabens are a group of EDCs commonly found in personal care products, foods, and pharmaceuticals. Systemic exposure to parabens has been confirmed by the ubiquitous detection of parabens in human blood and urine samples. Although evidence from in vivo and epidemiological studies linking parabens exposure to breast cancer is limited, the current evidence suggests that parabens may negatively interfere with some endocrine and intracrine targets relevant to breast carcinogenesis. So far, most studies have focused on a single paraben's effects and the direct modulating effects on estrogen receptors or the androgen receptor in vitro. Recent studies have revealed that parabens can modulate local estrogen-converting enzymes, 17β-hydroxysteroid dehydrogenase 1 and 2 and increase local estrogen levels. Also, parabens can crosstalk with the human epidermal growth factor receptor 2 (HER2) pathway and work with ER signaling to increase pro-oncogenic c-Myc expression in ER+/HER2+ breast cancer cells. Future studies investigating paraben mixtures and their crosstalk with other EDCs or signaling pathways both in vitro and in vivo in the context of breast cancer development are warranted.
Collapse
Affiliation(s)
- Emily Hager
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA;
| | - Jiangang Chen
- Department of Public Health, University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA;
| |
Collapse
|
23
|
Baicalin Inhibits EMT through PDK1/AKT Signaling in Human Nonsmall Cell Lung Cancer. JOURNAL OF ONCOLOGY 2021; 2021:4391581. [PMID: 34868313 PMCID: PMC8639265 DOI: 10.1155/2021/4391581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 11/17/2022]
Abstract
Background Baicalin is a naturally occurring compound with anticancer, antioxidant, and anti-inflammatory properties. However, the mechanism underlying its anticancer activity on nonsmall cell lung cancer (NSCLC) remains unclear. Methods The effects of baicalin on the progression and metastasis of experimental NSCLC cell lines were studied in vitro and in vivo. Wound-healing and transwell assays were performed to evaluate the potency of baicalin and the motility and migration ability of NCI-H460 cells. Immunofluorescence assay, western blot assay, and immunohistochemistry test were conducted to investigate the inhibiting effect of baicalin on the epithelial-mesenchymal transition (EMT) of NSCLC. Results Baicalin inhibited the proliferation and migration of NCI-H446 human NSCLC cells in a dose-dependent manner, reduced the expression levels of phospho-3-phosphoinositide-dependent protein kinase 1 (p-PDK1) and phosphor-serine/threonine-protein kinase (p-AKT), reversed the levels of EMT markers, and inhibited the migration of NSCLC cells. Conclusions Baicalin impedes EMT by inhibiting the PDK1/AKT pathway in human NSCLC and thus may be an effective alternative treatment for carcinoma and a new candidate antimetastasis drug.
Collapse
|
24
|
Yu Y, Wang W, Lu W, Chen W, Shang A. Inhibin β-A (INHBA) induces epithelial-mesenchymal transition and accelerates the motility of breast cancer cells by activating the TGF-β signaling pathway. Bioengineered 2021; 12:4681-4696. [PMID: 34346300 PMCID: PMC8806747 DOI: 10.1080/21655979.2021.1957754] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022] Open
Abstract
Accumulating evidence indicates that INHBA (Inhibin β-A, a member of the TGF-β superfamily) functions as an oncogene in cancer progression. However, little is known as to how INHBA regulates the progression and aggressiveness of breast cancer (BC). This study explored the function and underlying mechanism of INHBA in epithelial-mesenchymal transition (EMT) of BC cells. INHBA expression in BC cell lines was measured using RT-qPCR and Western blot. The would-healing and transwell migration assays were used to investigate the effect of INHBA overexpression or silencing on BC cell motility. Moreover, the expression levels of EMT-related genes were quantified after overexpressing or silencing of INHBA. Based on published dataset, INHBA was significantly upregulated in BC tissues compared to the adjacent normal tissues. A higher level of INHBA expression was also correlated with a poor survival in BC patients. In addition, in vitro study showed that INHBA played an indispensable role in promoting BC cell proliferation and invasion. Mechanistically, INHBA induced epithelial-mesenchymal transition (EMT) and accelerated the motility of BC cells by activating TGF-β-regulated genes. In conclusion, INHBA plays a functional role in supporting EMT phenotype of BC cells, and it may serve as a diagnostic biomarker and a potential therapeutic target for BC treatment.
Collapse
Affiliation(s)
- Yingying Yu
- Department of Obstetrics and Gynecology, the International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Weiwei Wang
- Department of Pathology, Tinghu District People’s Hospital of Yancheng City, Yancheng, Jiangsu, China
| | - Wenying Lu
- Department of Pathology, Tinghu District People’s Hospital of Yancheng City, Yancheng, Jiangsu, China
| | - Wei Chen
- Department of Emergency, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Anquan Shang
- Department of Laboratory Medicine, Shanghai Tongji Hospital, Shanghai, China
| |
Collapse
|
25
|
Gao L, Feng Y, Ge C, Xu X, Wang S, Li X, Zhang K, Wang C, Dai F, Xie S. Identification of molecular anti-metastasis mechanisms of lycorine in colorectal cancer by RNA-seq analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153530. [PMID: 33761445 DOI: 10.1016/j.phymed.2021.153530] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignancies worldwide. Metastasis is the major cause of death in patients with CRC. Lycorine, the phenanthridine alkaloid most commonly found in spp of the Amaryllidaceae family, has shown promising anticancer activities with minor side effects. However, the effects and the detailed mechanism of lycorine against metastasis of CRC remains unclear. STUDY DESIGN/METHODS The purpose of this study was to investigate the effects of lycorine on CRC and characterize the molecular mechanisms observed in lycorine-treated CRC cells using RNA-sequencing. MTT assay, colony formation assay, acridine orange/ethidium bromide (AO/EB) staining and Annexin V-FITC/Propidium iodide (PI) staining were conducted to examine the effects of lycorine on cell proliferation and apoptosis in CRC cells. RNA sequencing, real-time PCR assays and western blot were performed. Migration and invasion abilities of lycorine-treated CRC cells were investigated by wound healing and transwell invasion assays. The mouse CRC lung metastasis model was established and was used to detect the effect of lycorine on CRC in vivo. RESULTS Our results demonstrated that lycorine inhibited the proliferation and colony formation of CRC cells in a concentration-dependent manner. AO/EB staining and Annexin V-FITC/PI staining showed that lycorine induced apoptosis in a concentration-dependent manner. Lycorine also reduced lung metastasis of CRC in vivo. Moreover, transcriptomic analysis suggested that lycorine regulated the expression of 3556 genes. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway was implicated according to the differentially expressed genes (DEGs), and multiple pathways including those of mitogen-activated protein kinase (MAPK), relaxin, Ras, phosphatidylinositol 3‑kinase (PI3K)-protein kinase B (Akt) and Wnt/β-catenin were selected by functional enrichment analyses. Furthermore, based on transcriptomic analysis, we found that the tumor necrosis factor (TNF) pathway and endoplasmic reticulum stress were responsible for lycorine-induced apoptosis. CONCLUSIONS These results obtained in this study demonstrated that lycorine has the potential to suppress CRC in vitro and in vivo through the lycorine-regulated multiple signaling pathways.
Collapse
Affiliation(s)
- Lei Gao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng 475004, Henan, China; Joint International Research Laboratory of Food & Medicine Resource Function, Henan University, Kaifeng, 475004, Henan, China
| | - Yongli Feng
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng 475004, Henan, China
| | - Chaochao Ge
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng 475004, Henan, China
| | - Xiaojuan Xu
- School of Pharmacy, Henan University, Kaifeng 475004, Henan, China
| | - Senzhen Wang
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng 475004, Henan, China; School of Life Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Xinna Li
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng 475004, Henan, China
| | - Kemeng Zhang
- School of Life Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Chaojie Wang
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng 475004, Henan, China
| | - Fujun Dai
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng 475004, Henan, China; School of Life Sciences, Henan University, Kaifeng, 475004, Henan, China.
| | - Songqiang Xie
- School of Pharmacy, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
26
|
Zhao G, Zhang W, Dong P, Watari H, Guo Y, Pfeffer LM, Tigyi G, Yue J. EIF5A2 controls ovarian tumor growth and metastasis by promoting epithelial to mesenchymal transition via the TGFβ pathway. Cell Biosci 2021; 11:70. [PMID: 33827661 PMCID: PMC8025533 DOI: 10.1186/s13578-021-00578-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Epithelial to mesenchymal transition (EMT) contributes to tumor metastasis and chemoresistance. Eukaryotic initiation factor 5A2 (EIF5A2) is highly expressed in a variety of human cancers but rarely expressed in normal tissues. While EIF5A2 has oncogenic activity in several cancers and contributes to tumor metastasis, its role in ovarian cancer is unknown. In this study, we investigate whether EIF5A2 contributes to ovarian tumor metastasis by promoting EMT. METHODS To investigate the role of EIF5A2, we knocked out (KO) EIF5A2 using lentiviral CRISPR/Cas9 nickase in high invasive SKOV3 and OVCAR8 cells and overexpressed EIF5A2 in low invasive OVCAR3 cells using lentiviral vector. Cell proliferation, migration and invasion was examined in vitro ovarian cancer cells and tumor metastasis was evaluated in vivo using orthotopic ovarian cancer mouse models. RESULTS Here we report that EIF5A2 is highly expressed in ovarian cancers and associated with patient poor survival. Lentiviral CRISPR/Cas9 nickase vector mediated knockout (KO) of EIF5A2 inhibits epithelial to mesenchymal transition (EMT) in SKOV3 and OVCAR8 ovarian cancer cells that express high levels of EIF5A2. In contrast, overexpression of EIF5A2 promotes EMT in OVCAR3 epithelial adenocarcinoma cells that express relatively low EIF5A2 levels. KO of EIF5A2 in SKOV3 and OVCAR8 cells inhibits ovarian cancer cell migration and invasion, while its overexpression promotes cell migration and invasion in OVCAR3 adenocarcinoma cells. We further demonstrate that EIF5A2 promotes EMT by activating the TGFβ pathway and KO of EIF5A2 inhibits ovarian tumor growth and metastasis in orthotopic ovarian cancer mouse models. CONCLUSION Our results indicate that EIF5A2 is an important controller of ovarian tumor growth and metastasis by promoting EMT and activating the TGFβ pathway.
Collapse
Affiliation(s)
- Guannan Zhao
- Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Wenjing Zhang
- Department of Genetics, Genomics & Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638 Japan
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638 Japan
| | - Yuqi Guo
- People′s Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Lawrence M. Pfeffer
- Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Gabor Tigyi
- Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
- Center for Cancer Research, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163 USA
| |
Collapse
|
27
|
Peng F, Tang H, Du J, Chen J, Peng C. Isoliquiritigenin Suppresses EMT-Induced Metastasis in Triple-Negative Breast Cancer through miR-200c/C-JUN/
β
-Catenin. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:505-523. [PMID: 33641651 DOI: 10.1142/s0192415x21500233] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Triple-negative breast cancer (TNBC) is the subtype of breast cancer with more aggressive growth and metastasis and without efficient therapies. Hence, it is worthwhile to search for potential effective drug candidates. According to our previous study, isoliquiritigenin (ISL) exerted a potent anticancer effect on breast cancer proliferation. Its effect on TNBC growth, metastasis and mechanism deserves further investigation. In this study, PCR array screened a significant increase of miR-200c in BT-549 and MDA-MB-231 cells after ISL treatment, and ISH exerted that miR-200c was expressed at a low level in breast cancer tissue of patients. We also found that ISL could up-regulate miR-200c, resulting in the inhibition of epithelial-mesenchymal transition. Meanwhile, ISL could inhibit metastasis and tumor growth in nude mice models through the increase of miR-200c. Further study displayed that ISL decreased c-Jun expression through the increase of miR-200c. Interestingly, we also detected that ISL might increase miR-200c expression through the demethylation of miR-200c promoter region. These findings indicated that ISL could be potentially developed as a novel drug candidate for TNBC in microRNA-based cancer therapies.
Collapse
Affiliation(s)
- Fu Peng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional, Chinese Medicine, Chengdu, P. R. China
- West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | - Hailin Tang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P. R. China
| | - Junrong Du
- West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional, Chinese Medicine, Chengdu, P. R. China
| |
Collapse
|
28
|
Kong D, Zhou H, Neelakantan D, Hughes CJ, Hsu JY, Srinivasan RR, Lewis MT, Ford HL. VEGF-C mediates tumor growth and metastasis through promoting EMT-epithelial breast cancer cell crosstalk. Oncogene 2021; 40:964-979. [PMID: 33299122 PMCID: PMC7867573 DOI: 10.1038/s41388-020-01539-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 10/07/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
It is well established that a subset of cells within primary breast cancers can undergo an epithelial-to-mesenchymal transition (EMT), although the role of EMT in metastasis remains controversial. We previously demonstrated that breast cancer cells that had undergone an oncogenic EMT could increase metastasis of neighboring cancer cells via non-canonical paracrine-mediated activation of GLI activity that is dependent on SIX1 expression in the EMT cancer cells. However, the mechanism by which these SIX1-expressing EMT cells activate GLI signaling remained unclear. In this study, we demonstrate a novel mechanism for activation of GLI-mediated signaling in epithelial breast tumor cells via EMT cell-induced production and secretion of VEGF-C. We show that VEGF-C, secreted by breast cancer cells that have undergone an EMT, promotes paracrine-mediated increases in proliferation, migration, and invasion of epithelial breast cancer cells, via non-canonical activation of GLI-signaling. We further show that the aggressive phenotypes, including metastasis, imparted by EMT cells on adjacent epithelial cancer cells can be disrupted by either inhibiting VEGF-C in EMT cells or by knocking down NRP2, a receptor which interacts with VEGF-C, in neighboring epithelial cancer cells. Interrogation of TCGA and GEO public datasets supports the relevance of this pathway in human breast cancer, demonstrating that VEGF-C strongly correlates with activation of Hedgehog signaling and EMT in the human disease. Our study suggests that the VEGF-C/NRP2/GLI axis is a novel and conserved paracrine means by which EMT cells enhance metastasis, and provides potential targets for therapeutic intervention in this heterogeneous disease.
Collapse
Affiliation(s)
- Deguang Kong
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hengbo Zhou
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Deepika Neelakantan
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Connor J Hughes
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica Y Hsu
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Program in Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
29
|
Singh N, Mishra A, Sahu DK, Jain M, Shyam H, Tripathi RK, Shankar P, Kumar A, Alam N, Jaiswal R, Kumar S. Comprehensive Characterization of Stage IIIA Non-Small Cell Lung Carcinoma. Cancer Manag Res 2020; 12:11973-11988. [PMID: 33244273 PMCID: PMC7685366 DOI: 10.2147/cmar.s279974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/16/2020] [Indexed: 12/21/2022] Open
Abstract
Introduction Heterogeneity of non-small cell lung carcinoma (NSCLC) among patients is currently not well studied. Pathologic markers and staging systems have not been a precise predictor of the prognosis of an individual patient. Hence, we hypothesize to develop a transcript-based signature to categorize stage IIIA-NSCLC in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), plus identify markers that could indicate the prognosis of the disease. Methods Human Transcriptome Array 2.0 (HTA) and NanoString nCounter® platform were used for high-throughput gene-expression profiling. Initially, we profiled stage IIIA-NSCLC through HTA and validated through NanoString. Additionally, two metastatic markers SPP1 and CDH2 were validated in 47 NSCLC stage IIIA samples through real-time PCR. Results We observed distinct gene clusters in LUAD and LUSC with down-regulation of six genes and up-regulation of 57 genes through HTA. Ninety-six transcripts were randomly selected after analyzing HTA data and validated on the NanoString platform. We found 40 differentially expressed transcripts that categorized NSCLC into LUAD and LUSC. SPP1 is significantly overexpressed (4.311±1.27 fold in LUAD and 13.41±3.82 fold in LUSC compared to control), and the CDH2 transcript was significantly overexpressed (11.53 ± 4.027-fold compared to control) only in LUSC. Discussion These markers enable us to categorize stage IIIA NSCLC into LUAD and LUSC plus these markers may be helpful to understand the pathophysiology of NSCLC. However, more data required to make these findings useful in general clinical practice.
Collapse
Affiliation(s)
- Neetu Singh
- Department of Centre for Advanced Research, King George's Medical University, Lucknow, 226003, India
| | - Archana Mishra
- Department of Surgery, King George's Medical University, Lucknow 226003, India
| | - Dinesh Kumar Sahu
- Department of Centre for Advanced Research, King George's Medical University, Lucknow, 226003, India
| | - Mayank Jain
- Department of Centre for Advanced Research, King George's Medical University, Lucknow, 226003, India
| | - Hari Shyam
- Department of Centre for Advanced Research, King George's Medical University, Lucknow, 226003, India
| | - Ratnesh Kumar Tripathi
- Department of Centre for Advanced Research, King George's Medical University, Lucknow, 226003, India
| | - Pratap Shankar
- Department of Centre for Advanced Research, King George's Medical University, Lucknow, 226003, India
| | - Anil Kumar
- Department of Centre for Advanced Research, King George's Medical University, Lucknow, 226003, India
| | - Nawazish Alam
- Department of Centre for Advanced Research, King George's Medical University, Lucknow, 226003, India
| | - Riddhi Jaiswal
- Department of Pathology, King George's Medical University, Lucknow 226003, India
| | - Shailendra Kumar
- Department of Surgery, King George's Medical University, Lucknow 226003, India
| |
Collapse
|
30
|
Angius A, Cossu-Rocca P, Arru C, Muroni MR, Rallo V, Carru C, Uva P, Pira G, Orrù S, De Miglio MR. Modulatory Role of microRNAs in Triple Negative Breast Cancer with Basal-Like Phenotype. Cancers (Basel) 2020; 12:E3298. [PMID: 33171872 PMCID: PMC7695196 DOI: 10.3390/cancers12113298] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 12/16/2022] Open
Abstract
Development of new research, classification, and therapeutic options are urgently required due to the fact that TNBC is a heterogeneous malignancy. The expression of high molecular weight cytokeratins identifies a biologically and clinically distinct subgroup of TNBCs with a basal-like phenotype, representing about 75% of TNBCs, while the remaining 25% includes all other intrinsic subtypes. The triple negative phenotype in basal-like breast cancer (BLBC) makes it unresponsive to endocrine therapy, i.e., tamoxifen, aromatase inhibitors, and/or anti-HER2-targeted therapies; for this reason, only chemotherapy can be considered an approach available for systemic treatment even if it shows poor prognosis. Therefore, treatment for these subgroups of patients is a strong challenge for oncologists due to disease heterogeneity and the absence of unambiguous molecular targets. Dysregulation of the cellular miRNAome has been related to huge cellular process deregulations underlying human malignancy. Consequently, epigenetics is a field of great promise in cancer research. Increasing evidence suggests that specific miRNA clusters/signatures might be of clinical utility in TNBCs with basal-like phenotype. The epigenetic mechanisms behind tumorigenesis enable progress in the treatment, diagnosis, and prevention of cancer. This review intends to summarize the epigenetic findings related to miRNAome in TNBCs with basal-like phenotype.
Collapse
Affiliation(s)
- Andrea Angius
- Institute of Genetic and Biomedical Research (IRGB), CNR, Cittadella Universitaria di Cagliari, 09042 Monserrato, Italy;
| | - Paolo Cossu-Rocca
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (P.C.-R.); (M.R.M.)
- Department of Diagnostic Services, “Giovanni Paolo II” Hospital, ASSL Olbia-ATS Sardegna, 07026 Olbia, Italy
| | - Caterina Arru
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.A.); (C.C.); (G.P.)
| | - Maria Rosaria Muroni
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via P. Manzella, 4, 07100 Sassari, Italy; (P.C.-R.); (M.R.M.)
| | - Vincenzo Rallo
- Institute of Genetic and Biomedical Research (IRGB), CNR, Cittadella Universitaria di Cagliari, 09042 Monserrato, Italy;
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.A.); (C.C.); (G.P.)
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.A.); (C.C.); (G.P.)
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Piscina Manna, 09010 Pula, CA, Italy;
| | - Giovanna Pira
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.A.); (C.C.); (G.P.)
| | - Sandra Orrù
- Department of Pathology, “A. Businco” Oncologic Hospital, ASL Cagliari, 09121 Cagliari, Italy;
| | - Maria Rosaria De Miglio
- Institute of Genetic and Biomedical Research (IRGB), CNR, Cittadella Universitaria di Cagliari, 09042 Monserrato, Italy;
| |
Collapse
|
31
|
Qi Y, Liu J, Chao J, Scheuerman MP, Rahimi SA, Lee LY, Li S. PTEN suppresses epithelial-mesenchymal transition and cancer stem cell activity by downregulating Abi1. Sci Rep 2020; 10:12685. [PMID: 32728066 PMCID: PMC7391766 DOI: 10.1038/s41598-020-69698-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
The epithelial–mesenchymal transition (EMT) is an embryonic program frequently reactivated during cancer progression and is implicated in cancer invasion and metastasis. Cancer cells can also acquire stem cell properties to self-renew and give rise to new tumors through the EMT. Inactivation of the tumor suppressor PTEN has been shown to induce the EMT, but the underlying molecular mechanisms are less understood. In this study, we reconstituted PTEN-deficient breast cancer cells with wild-type and mutant PTEN, demonstrating that restoration of PTEN expression converted cancer cells with mesenchymal traits to an epithelial phenotype and inhibited cancer stem cell (CSC) activity. The protein rather than the lipid phosphatase activity of PTEN accounts for the reversal of the EMT. PTEN dephosphorylates and downregulates Abi1 in breast cancer cells. Gain- and loss-of-function analysis indicates that upregulation of Abi1 mediates PTEN loss-induced EMT and CSC activity. These results suggest that PTEN may suppress breast cancer invasion and metastasis via dephosphorylating and downregulating Abi1.
Collapse
Affiliation(s)
- Yanmei Qi
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, 125 Paterson Street, MEB-687, New Brunswick, NJ, 08093, USA
| | - Jie Liu
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, 125 Paterson Street, MEB-687, New Brunswick, NJ, 08093, USA
| | - Joshua Chao
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, 125 Paterson Street, MEB-687, New Brunswick, NJ, 08093, USA
| | - Mark P Scheuerman
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, 125 Paterson Street, MEB-687, New Brunswick, NJ, 08093, USA
| | - Saum A Rahimi
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, 125 Paterson Street, MEB-687, New Brunswick, NJ, 08093, USA
| | - Leonard Y Lee
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, 125 Paterson Street, MEB-687, New Brunswick, NJ, 08093, USA
| | - Shaohua Li
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, 125 Paterson Street, MEB-687, New Brunswick, NJ, 08093, USA.
| |
Collapse
|
32
|
Wang Y, Liao R, Chen X, Ying X, Chen G, Li M, Dong C. Twist-mediated PAR1 induction is required for breast cancer progression and metastasis by inhibiting Hippo pathway. Cell Death Dis 2020; 11:520. [PMID: 32647142 PMCID: PMC7347637 DOI: 10.1038/s41419-020-2725-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 06/24/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer is considered to be the most prevalent cancer in women worldwide, and metastasis is the primary cause of death. Protease-activated receptor 1 (PAR1) is a GPCR family member involved in the invasive and metastatic processes of cancer cells. However, the functions and underlying mechanisms of PAR1 in breast cancer remain unclear. In this study, we found that PAR1 is highly expressed in high invasive breast cancer cells, and predicts poor prognosis in ER-negative and high-grade breast cancer patients. Mechanistically, Twist transcriptionally induces PAR1 expression, leading to inhibition of Hippo pathway and activation of YAP/TAZ; Inhibition of PAR1 suppresses YAP/TAZ-induced epithelial-mesenchymal transition (EMT), invasion, migration, cancer stem cell (CSC)-like properties, tumor growth and metastasis of breast cancer cells in vitro and in vivo. These findings suggest that PAR1 acts as a direct transcriptionally target of Twist, can promote EMT, tumorigenicity and metastasis by controlling the Hippo pathway; this may lead to a potential therapeutic target for treating invasive breast cancer.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute of Integrative Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruocen Liao
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingyu Chen
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuhua Ying
- Cancer Institute of Integrative Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Guanping Chen
- Cancer Institute of Integrative Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Mingqian Li
- Cancer Institute of Integrative Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (breast center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
33
|
Veloso ES, Gonçalves INN, Silveira TL, Espirito Santo JT, Figueiredo LV, Varaschin MS, Cassali GD, Del Puerto HL, Ferreira E. ZEB and Snail expression indicates epithelial-mesenchymal transition in canine melanoma. Res Vet Sci 2020; 131:7-14. [PMID: 32278962 DOI: 10.1016/j.rvsc.2020.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 11/15/2022]
Abstract
Melanoma progression is associated with the epithelial-mesenchymal transition (EMT) when tumor cells reduce E-cadherin and increase N-cadherin expression resulting in an escape from the microenvironment via loss of cellular adhesion and gain of motility. Transcription factor proteins Snail and ZEB trigger EMT by repression of epithelial markers and activation of mesenchymal properties. This study evaluated E-cadherin, N-cadherin, Snail, ZEB1 and ZEB2 expression by IHC and investigated their relationship with morphological characteristics in cutaneous and oral canine melanoma. Results from melanoma cases demonstrated E-cadherin expression in 45% (9/20) of oral and 58% (22/38) of cutaneous tumors, while N-cadherin expression was observed in 95% (18/19) of oral and 92% (34/37) of cutaneous melanoma. Cytoplasmic and nuclear N-cadherin expression was positively correlated with ZEB1 expression, while the cell membrane N-cadherin expression was positively correlated with ZEB2. In addition, an increase in nuclear N-cadherin expression was associated with reduced Snail expression in cutaneous melanoma and an increase in Snail expression in oral melanoma, indicating that the correlation between N-cadherin and Snail expression is coincident with tumor location. Our data suggest that ZEB family protein is associated with N-cadherin translocation from cell membrane to the cytoplasm and nuclei, and may act as important transcription factors of EMT regulation in canine melanoma.
Collapse
Affiliation(s)
- Emerson Soares Veloso
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, MG, Brazil
| | | | - Tatiany Luiza Silveira
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, MG, Brazil
| | | | - Larissa Vieira Figueiredo
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, MG, Brazil
| | | | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, MG, Brazil
| | - Helen Lima Del Puerto
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, MG, Brazil
| | - Enio Ferreira
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, MG, Brazil.
| |
Collapse
|
34
|
Peng F, Xiong L, Peng C. (-)-Sativan Inhibits Tumor Development and Regulates miR-200c/PD-L1 in Triple Negative Breast Cancer Cells. Front Pharmacol 2020; 11:251. [PMID: 32231566 PMCID: PMC7082844 DOI: 10.3389/fphar.2020.00251] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 02/24/2020] [Indexed: 01/17/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) in cancer cells could convert epithelial-like cells to mesenchymal-like cells, resulting in the increased capacity of migration and invasion of cancer cells, and is an essential step in triple negative breast cancer (TNBC) development. Recent reports exert that these EMT-activated TNBC cells are more resistant to immune attacks, with high levels of programmed death ligand1 (PD-L1). Hence, it is worthwhile to find an effective approach in inhibiting EMT-activated TNBC cells. (-)-Sativan (SA) is a naturally isolated isoflavane and could be isolated from Spatholobus suberectus, a common traditional Chinese medicine used for breast cancer treatment. It was the first time that SA exerted anti-cancer effects on breast cancer cells, according to our study. In this study, SA displayed a significant inhibitory effect on the proliferation of TNBC cells by inducing apoptosis. SA increased Bax expression, and decreased Bcl-2 protein levels. SA inhibited cell migration and invasion of MDA-MB-231 and BT-549 cells. SA could decrease N-cadherin, Snail, Vimentin, and PD-L1 expression. SA increased miR-200c expression, and decreased PD-L1 expression. Luciferase assay showed that miR-200c directly targeted PD-L1. SA promoted tumor cell susceptibility to CTL-mediated lysis. Further study confirmed that SA could inhibit PD-L1 expression and EMT by up-regulating miR-200c. In vivo results displayed that SA could also inhibit tumor volumes and weights. These findings indicate that SA exerts an inhibitory effect on TNBC cell proliferation, migration, invasion, and tumor gtrowth, and partly provide evidence for the anti-breast cancer effect of Spatholobus suberectus Dunn in TNBC therapy.
Collapse
Affiliation(s)
- Fu Peng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, China.,Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liang Xiong
- Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
35
|
Gholami MD, Falak R, Heidari S, Khoshmirsafa M, Kazemi MH, Zarnani AH, Safari E, Tajik N, Kardar GA. A Truncated Snail1 Transcription Factor Alters the Expression of Essential EMT Markers and Suppresses Tumor Cell Migration in a Human Lung Cancer Cell Line. Recent Pat Anticancer Drug Discov 2020; 14:158-169. [PMID: 31131753 DOI: 10.2174/1574892814666190527111429] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 05/08/2019] [Accepted: 05/24/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Epithelial-to-Mesenchymal Transition (EMT) is necessary for metastasis. Zinc- finger domain-containing transcription factors, especially Snail1, bind to E-box motifs and play a crucial role in the induction and regulation of EMT. OBJECTIVE We hypothesized if C-terminal region of Snail1 (CSnail1) may competitively bind to E-box and block cancer metastasis. METHODS The CSnail1 gene coding sequence was inserted into the pIRES2-EGFP vector. Following transfection of A549 cells with the designed construct, EMT was induced with TGF-β1 and the expression of essential EMT markers was evaluated by real-time PCR and immunoblotting. We also monitored cell migration. RESULTS CSnail1 inhibited TGF-β1-induced N-cadherin and vimentin mRNA expression and increased β-catenin expression in transfected TGF-β1-treated A549 cells. A similar finding was obtained in western blotting. CSnail1 also blocked the migration of transfected cells in the scratch test. CONCLUSION Transfection of A549 cells with CSnail1 alters the expression of essential EMT markers and consequently suppresses tumor cell migration. These findings confirm the capability of CSnail1 in EMT blocking and in parallel to current patents could be applied as a novel strategy in the prevention of metastasis.
Collapse
Affiliation(s)
- Mohammad Davoodzadeh Gholami
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sahel Heidari
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Khoshmirsafa
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad H Kazemi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Elaheh Safari
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nader Tajik
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Gholam A Kardar
- Immunology Asthma & Allergy Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Guo Y, Fan Y, Pei X. Fangjihuangqi Decoction inhibits MDA-MB-231 cell invasion in vitro and decreases tumor growth and metastasis in triple-negative breast cancer xenografts tumor zebrafish model. Cancer Med 2020; 9:2564-2578. [PMID: 32037729 PMCID: PMC7131862 DOI: 10.1002/cam4.2894] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 01/11/2020] [Accepted: 01/19/2020] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a basal-like cancer which is considered to be more intrusive, have a poorer prognosis and chemoresistance. TNBC is characterized by the presence of epithelial to mesenchymal transition (EMT) that plays a major role in the progression of the cancer. In the present study, we first use a classic prescription of Chinese medicine Fangjihuangqi Decoction to treat TGFβ1-induced MDA-MB-231 cells in vitro. Our data showed that TGFβ1-induced MDA-MB-231 cell morphology change, promoted MDA-MB 231 invasion, increased Vimentin expression, and decreased E-cadherin expression. Further, Fangjihuangqi Decoction-medicated serum (FHS) treated both MDA-MB 231 cells and TGFβ1-induced MDA-MB-231 cells. Results showed that Fangjihuangqi Decoction could inhibit cell proliferation, reduce cell invasion, increase E-cadherin expression, and decrease EMT markers. Secondly, we established a xenograft tumor zebrafish model to assess Fangjihuangqi Decoction inhibition of cancer cell proliferation and invasion. Our results indicated that Fangjihuangqi Decoction could inhibit tumor growth, restrain the sprouts number of tumor neovascularization, and reduce the length of tumor neoplastic lymphatics by increasing E-cadherin expression and decreasing EMT markers in TNBC xenograft tumor zebrafish model. Overall, our studies provide evidences that Fangjihuangqi Decoction could inhibit TNBC, reverse EMT, and contribute to antimetastasis by increasing E-cadherin expression and decreasing EMT markers, which provide an experimental basis for clinical application of Fangjihuangqi Decoction on TNBC treatment.
Collapse
Affiliation(s)
- Yubo Guo
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yingyi Fan
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohua Pei
- Beijing University of Chinese Medicine Fangshan Traditional Medical Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
37
|
Wang Y, Dong C, Zhou BP. Metabolic reprogram associated with epithelial-mesenchymal transition in tumor progression and metastasis. Genes Dis 2019; 7:172-184. [PMID: 32215287 PMCID: PMC7083713 DOI: 10.1016/j.gendis.2019.09.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 09/08/2019] [Accepted: 09/27/2019] [Indexed: 02/09/2023] Open
Abstract
Epithelial-mesenchymal Transition (EMT) is a de-differentiation program that imparts tumor cells with the phenotypic and cellular plasticity required for drug resistance, metastasis, and recurrence. This dynamic and reversible events is governed by a network of EMT-transcription factors (EMT-TFs) through epigenetic regulation. Many chromatin modifying-enzymes utilize metabolic intermediates as cofactors or substrates; this suggests that EMT is subjected to the metabolic regulation. Conversely, EMT rewires metabolic program to accommodate cellular changes during EMT. Here we summarize the latest findings regarding the epigenetic regulation of EMT, and discuss the mutual interactions among metabolism, epigenetic regulation, and EMT. Finally, we provide perspectives of how this interplay contributes to cellular plasticity, which may result in the clinical manifestation of tumor heterogeneity.
Collapse
Affiliation(s)
- Yifan Wang
- Cancer Institute of Integrative Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310012, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, Department of Surgical Oncology (Breast Center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY, 40506, USA
| |
Collapse
|
38
|
Li CJ, Chu PY, Yiang GT, Wu MY. The Molecular Mechanism of Epithelial-Mesenchymal Transition for Breast Carcinogenesis. Biomolecules 2019; 9:biom9090476. [PMID: 31514467 PMCID: PMC6770718 DOI: 10.3390/biom9090476] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 12/23/2022] Open
Abstract
The transforming growth factor-β (TGF-β) signaling pathway plays multiple regulatory roles in the tumorigenesis and development of cancer. TGF-β can inhibit the growth and proliferation of epithelial cells and induce apoptosis, thereby playing a role in inhibiting breast cancer. Therefore, the loss of response in epithelial cells that leads to the inhibition of cell proliferation due to TGF-β is a landmark event in tumorigenesis. As tumors progress, TGF-β can promote tumor cell invasion, metastasis, and drug resistance. At present, the above-mentioned role of TGF-β is related to the interaction of multiple signaling pathways in the cell, which can attenuate or abolish the inhibition of proliferation and apoptosis-promoting effects of TGF-β and enhance its promotion of tumor progression. This article focuses on the molecular mechanisms through which TGF-β interacts with multiple intracellular signaling pathways in tumor progression and the effects of these interactions on tumorigenesis.
Collapse
Affiliation(s)
- Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
| | - Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| |
Collapse
|
39
|
Christowitz C, Davis T, Isaacs A, van Niekerk G, Hattingh S, Engelbrecht AM. Mechanisms of doxorubicin-induced drug resistance and drug resistant tumour growth in a murine breast tumour model. BMC Cancer 2019; 19:757. [PMID: 31370818 PMCID: PMC6670209 DOI: 10.1186/s12885-019-5939-z] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/15/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Doxorubicin is currently the most effective chemotherapeutic drug used to treat breast cancer. It has, however, been shown that doxorubicin can induce drug resistance resulting in poor patient prognosis and survival. Studies reported that the interaction between signalling pathways can promote drug resistance through the induction of proliferation, cell cycle progression and prevention of apoptosis. The aim of this study was therefore to determine the effects of doxorubicin on apoptosis signalling, autophagy, the mitogen-activated protein kinase (MAPK)- and phosphoinositide 3-kinase (PI3K)/Akt signalling pathway, cell cycle control, and regulators of the epithelial-mesenchymal transition (EMT) process in murine breast cancer tumours. METHODS A tumour-bearing mouse model was established by injecting murine E0771 breast cancer cells, suspended in Hank's Balances Salt Solution and Corning® Matrigel® Basement Membrane Matrix, into female C57BL/6 mice. Fourty-seven mice were randomly divided into three groups, namely tumour control (received Hank's Balances Salt Solution), low dose doxorubicin (received total of 6 mg/ml doxorubicin) and high dose doxorubicin (received total of 15 mg/ml doxorubicin) groups. A higher tumour growth rate was, however, observed in doxorubicin-treated mice compared to the untreated controls. We therefore compared the expression levels of markers involved in cell death and survival signalling pathways, by means of western blotting and fluorescence-based immunohistochemistry. RESULTS Doxorubicin failed to induce cell death, by means of apoptosis or autophagy, and cell cycle arrest, indicating the occurrence of drug resistance and uncontrolled proliferation. Activation of the MAPK/ extracellular-signal-regulated kinase (ERK) pathway contributed to the resistance observed in treated mice, while no significant changes were found with the PI3K/Akt pathway and other MAPK pathways. Significant changes were also observed in cell cycle p21 and DNA replication minichromosome maintenance 2 proteins. No significant changes in EMT markers were observed after doxorubicin treatment. CONCLUSIONS Our results suggest that doxorubicin-induced drug resistance and tumour growth can occur through the adaptive role of the MAPK/ERK pathway in an effort to protect tumour cells. Previous studies have shown that the efficacy of doxorubicin can be improved by inhibition of the ERK signalling pathway and thereby treatment failure can be overcome.
Collapse
Affiliation(s)
- Claudia Christowitz
- Department of Global Health, Faculty of Medicine and Health Sciences, African Cancer Institute, Stellenbosch University, Cape Town, 8000 South Africa
| | - Tanja Davis
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, 7600 South Africa
| | - Ashwin Isaacs
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, 7600 South Africa
| | - Gustav van Niekerk
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, 7600 South Africa
| | - Suzel Hattingh
- Department of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 8000 South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, 7600 South Africa
| |
Collapse
|
40
|
Dong J, Yang W, Han J, Cheng R, Guo X, Li L. Effect of dihydroartemisinin on epithelial-to-mesenchymal transition in canine mammary tumour cells. Res Vet Sci 2019; 124:240-247. [PMID: 30947110 DOI: 10.1016/j.rvsc.2019.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 03/23/2019] [Accepted: 03/24/2019] [Indexed: 01/18/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process by which epithelial cells acquire mesenchymal properties, generating metastases. Dihydroartemisinin (DHA) exhibits potent anti-cancer activities. CMTs is a potential suitable model for studies in human breast cancer research. In the present study, we separated DHA-untreated cells from CMTs cells to be a control group, the rest of the CMTs cells treated by DHA which were composed of three concentrations 5,10 as well as 20 μM. After that we cultivate the cells severally under the condition of 24, 48 and 72 h at 37 °C. CMTs cells were evaluated by Cell viability assay, Wound healing assay, Invasion assay and RT-PCR analysis for the expression of Slug, ZEB1, ZEB2 and Twist. Our results showed that DHA increased the inhibitory rate of CMTs cell and restrain TGF-β1-induced migration and invasion of cells in a time and concentration reliant manner efficaciously. Our result also show DHA inhibits the expression of Slug, ZEB1, ZEB2 and Twist at the mRNA in vitro, the media concentration DHA (10 μM) decreased the expression level of the Slug, ZEB1, ZEB2 mRNA significantly. In conclusion, DHA inhibits canine mammary tumours cells migration and invasiveness by regulating the expression of EMT-related genes.
Collapse
Affiliation(s)
- Jing Dong
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Wenhui Yang
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Jiaqi Han
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Rongjie Cheng
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Xin Guo
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Lin Li
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China.
| |
Collapse
|
41
|
Wang R, Zhao H, Zhang X, Zhao X, Song Z, Ouyang J. Metabolic Discrimination of Breast Cancer Subtypes at the Single-Cell Level by Multiple Microextraction Coupled with Mass Spectrometry. Anal Chem 2019; 91:3667-3674. [DOI: 10.1021/acs.analchem.8b05739] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Ruihua Wang
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hansen Zhao
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Xiaochao Zhang
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Xu Zhao
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Zhe Song
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jin Ouyang
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
42
|
Jin X, Wei Y, Liu Y, Lu X, Ding F, Wang J, Yang S. Resveratrol promotes sensitization to Doxorubicin by inhibiting epithelial-mesenchymal transition and modulating SIRT1/β-catenin signaling pathway in breast cancer. Cancer Med 2019; 8:1246-1257. [PMID: 30697969 PMCID: PMC6434195 DOI: 10.1002/cam4.1993] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/01/2018] [Accepted: 01/04/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is one of the leading fatal diseases for women worldwide who cannot have surgery typically have to rely on systemic chemotherapy to extend their survival. Doxorubicin (DOX) is one of the most commonly used chemotherapeutic agents against breast cancer, but acquired resistance to DOX can seriously impede the efficacy of chemotherapy, leading to poor prognosis and recurrences of cancer. Resveratrol (RES) is a phytoalexin with pharmacological antitumor properties, but its underlying mechanisms are not clearly understood in the treatment of DOX‐resistant breast cancer. We used cell viability assays, cell scratch tests, and transwell assays combined with Western blotting and immunofluorescent staining to evaluate the effects of RES on chemoresistance and the epithelial‐mesenchymal transitions (EMTs) in adriamycin‐resistant MCF7/ADR breast cancer cells, and to investigate its underlying mechanisms. The results showed that a treatment of RES combining with DOX effectively inhibited cell growth, suppressed cell migration, and promoted cell apoptosis. RES reversed EMT properties of MCF7/ADR cells by modulating the connection between SIRT1 and β‐catenin, which provides a hopeful therapeutic avenue to conquer DOX‐resistance and thereby prolong survival rates in breast cancer patients.
Collapse
Affiliation(s)
- Xiaoxia Jin
- Department of Pathology, Nantong Tumor Hospital, Nantong, China
| | - Yingze Wei
- Department of Pathology, Nantong Tumor Hospital, Nantong, China
| | - Yushan Liu
- Department of Pathology, Nantong Tumor Hospital, Nantong, China
| | - Xiaoyun Lu
- Department of Pathology, Nantong Tumor Hospital, Nantong, China
| | - Fei Ding
- Department of Pathology, Nantong Tumor Hospital, Nantong, China
| | - Jiatai Wang
- Department of Pathology, Nantong Tumor Hospital, Nantong, China
| | - Shuyun Yang
- Department of Pathology, Nantong Tumor Hospital, Nantong, China
| |
Collapse
|
43
|
Gamba CDO, Damasceno KA, Ferreira IC, Rodrigues MA, Gomes DA, Alves MR, Rocha RM, Lima AE, Ferreira E, Cassali GD. The investigation of transcriptional repression mediated by ZEB2 in canine invasive micropapillary carcinoma in mammary gland. PLoS One 2019; 14:e0209497. [PMID: 30645591 PMCID: PMC6333364 DOI: 10.1371/journal.pone.0209497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023] Open
Abstract
The E-cadherin loss has frequently been associated with transcriptional repression mediated by transcription factors, such as the Zinc Finger E-Box Binding Homeobox-2 (ZEB2). Invasive micropapillary carcinomas (IMPCs) of the breast are aggressive neoplasms frequently related to lymph node metastasis and poor overall survival. In the canine mammary gland, IMPCs has just been reported and, based on its behavioral similarity with the human IMPCs, appears to be a good spontaneous model to this human entity. This study aimed to evaluate the relationship between E-cadherin and ZEB2 in a spontaneous canine model of invasive micropapillary carcinoma of the mammary gland. The correlation among gene expression (ZEB2 and CDH1) and clinicopathological findings was also explored. Nineteen cases of IMPC of the canine mammary gland were obtained, protein and mRNA expression were investigated through immunohistochemistry and RNA In Situ Hybridization, respectively. To better understand the relationship between E-cadherin and ZEB2, immunofluorescence was performed in canine IMPCs. Immunohistochemically, most of IMPCs showed 1+ (14/19, 73.7%) for E-cadherin; and positivity for ZEB2 was diagnosed in 47.4% of the IMPCs. Regarding the RNA In Situ Hybridization (ISH), most of IMPCs showed 4+ and 0+ for E-cadherin (CDH1) and ZEB2 respectively. Through immunofluorescence, the first and second more frequent combinatorial group were E-cadherin+ZEB2- and E-cadherin+ZEB2+; neoplastic cells showing concomitantly weak expression for E-cadherin and positivity for ZEB2 were frequently observed. A negative correlation was observed between E-cadherin and progesterone receptor expression in IMPCs. Based on these results, canine mammary IMPCs show E-cadherin lost and, at times reveals nuclear positivity for the transcription factor ZEB2 that seems to exert transcriptional repression of the CDH1.
Collapse
Affiliation(s)
- Conrado de Oliveira Gamba
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Curso de Medicina Veterinária, Universidade do Oeste de Santa Catarina, Campus aproximado de Campos Novos, Campos Novos, Santa Catarina, Brazil
| | - Karine Araújo Damasceno
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Laboratório de Patologia Experimental, Instituto Gonçalo Moniz, Fiocruz, Salvador, Bahia, Brazil
| | - Izabel Cristina Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Michele Angela Rodrigues
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Dawidson Assis Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana Resende Alves
- International Center of Research in Cancer (CIPE), A.C. Camargo Cancer Center, São Paulo, São Paulo, Brazil
| | - Rafael Malagoli Rocha
- International Center of Research in Cancer (CIPE), A.C. Camargo Cancer Center, São Paulo, São Paulo, Brazil
| | - Alessandra Estrela Lima
- Departamento de Patologia e Clínicas, Escola de Medicina Veterinária, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Enio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Geovanni Dantas Cassali
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
44
|
Transfection of T-Box Transcription Factor BRACHYURY and SOX2 Synergistically Promote Self-Renewal and Invasive Phenotype in Oral Cancer Cells. Int J Mol Sci 2018; 19:ijms19113620. [PMID: 30453543 PMCID: PMC6275038 DOI: 10.3390/ijms19113620] [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] [Received: 10/08/2018] [Revised: 11/06/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022] Open
Abstract
Recent studies suggest that epithelial–mesenchymal transition (EMT) correlates with cancer metastasis. In addition, there is growing evidence of the association of EMT with cancer stem cells (CSCs). Recently, we showed that the T-box transcription factor BRACHYURY could be a strong regulator of EMT and the CSC phenotype, which were effectively suppressed by a BRACHYURY knockdown in an adenoid cystic carcinoma cell line. In this study, we further tested whether BRACHYURY is a regulator of cancer stemness by means of forced expression of BRACHYURY in oral cancer cell lines. BRACHYURY, SOX2, or both were stably transfected into oral carcinoma cell lines. We analysed these transfectants with respect to self-renewal phenotypes using a sphere-formation assay, and we assessed the expression levels of EMT markers and stem cell markers using real-time reverse transcription-polymerase chain reaction (RT-PCR). Cell migration and invasiveness in vitro were evaluated using a wound healing assay and a tumour cell dissemination assay, respectively. Forced expression of BRACHYURY or SOX2 slightly increased expression of EMT and stem cell markers and the self-renewal phenotype. The expression levels, however, were much lower compared to those of cancer stem cell-like cells. Forced co-expression of BRACHYURY and SOX2 strongly upregulated EMT and stem cell markers and the self-renewal phenotype. Cell migration and invasiveness in vitro were also remarkably enhanced. These synergistic effects increased expression levels of FIBRONECTIN, SNAIL, SLUG, ZEB1, and TGF-β2. In particular, the effects on FIBRONECTIN and TGF-β2 were significant. We found that BRACHYURY and SOX2 synergistically promote cancer stemness in oral cancer cells. This finding points to the importance of gene or protein networks associated with BRACHYURY and SOX2 in the development and maintenance of the CSC phenotype.
Collapse
|
45
|
Dittmer J. Breast cancer stem cells: Features, key drivers and treatment options. Semin Cancer Biol 2018; 53:59-74. [PMID: 30059727 DOI: 10.1016/j.semcancer.2018.07.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023]
Abstract
The current view is that breast cancer is a stem cell disease characterized by the existence of cancer cells with stem-like features and tumor-initiating potential. These cells are made responsible for tumor dissemination and metastasis. Common therapies by chemotherapeutic drugs fail to eradicate these cells and rather increase the pool of cancer stem cells in tumors, an effect that may increase the likelyhood of recurrence. Fifteen years after the first evidence for a small stem-like subpopulation playing a major role in breast cancer initiation has been published a large body of knowledge has been accumulated regarding the signaling cascades and proteins involved in maintaining stemness in breast cancer. Differences in the stem cell pool size and in mechanisms regulating stemness in the different breast cancer subtypes have emerged. Overall, this knowledge offers new approaches to intervene with breast cancer stem cell activity. New options are particularly needed for the treatment of triple-negative breast cancer subtype, which is particularly rich in cancer stem cells and is also the subtype for which specific therapies are still not available.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
46
|
Sun R, Xie HY, Qian JX, Huang YN, Yang F, Zhang FL, Shao ZM, Li DQ. FBXO22 Possesses Both Protumorigenic and Antimetastatic Roles in Breast Cancer Progression. Cancer Res 2018; 78:5274-5286. [PMID: 29945959 DOI: 10.1158/0008-5472.can-17-3647] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 11/16/2022]
Abstract
The molecular underpinnings behind malignant progression of breast cancer from a localized lesion to an invasive and ultimately metastatic disease are incompletely understood. Here, we report that F-box only protein 22 (FBXO22) plays a dual role in mammary tumorigenesis and metastasis. FBXO22 was upregulated in primary breast tumors and promoted cell proliferation and colony formation in vitro and xenograft tumorigenicity in vivo Surprisingly, FBXO22 suppressed epithelial-mesenchymal transition (EMT), cell motility, and invasiveness in vitro and metastatic lung colonization in vivo Clinical data showed that expression levels of FBXO22 were associated with favorable clinical outcomes, supporting the notion that metastasis, rather than primary cancer, is the major determinant of the mortality of patients with breast cancer. Mechanistic investigations further revealed that FBXO22 elicits its antimetastatic effects by targeting SNAIL, a master regulator of EMT and breast cancer metastasis, for ubiquitin-mediated proteasomal degradation in a glycogen synthase kinase 3β phosphorylation-dependent manner. Importantly, expression of SNAIL rescued FBXO22-mediated suppression of EMT, cell migration, and invasion. A patient-derived tryptophan-to-arginine mutation at residue 52 (W52R) within the F-box domain impaired FBXO22 binding to the SKP1-Cullin1 complex and blocked FBXO22-mediated SNAIL degradation, thus abrogating the ability of FBXO22 to suppress cell migration, invasion, and metastasis. Collectively, these findings uncover an unexpected dual role for FBXO22 in mammary tumorigenesis and metastatic progression and delineate the mechanism of an oncogenic mutation of FBXO22 in breast cancer progression.Significance: These findings highlight the paradoxical roles of FBXO22 in breast cancer, as it promotes breast tumor cell proliferation but prevents EMT and metastasis. Cancer Res; 78(18); 5274-86. ©2018 AACR.
Collapse
Affiliation(s)
- Rui Sun
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Yan Xie
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin-Xian Qian
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan-Ni Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fan Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang-Lin Zhang
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Min Shao
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Da-Qiang Li
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Raposo-Ferreira TMM, Brisson BK, Durham AC, Laufer-Amorim R, Kristiansen V, Puré E, Volk SW, Sorenmo K. Characteristics of the Epithelial-Mesenchymal Transition in Primary and Paired Metastatic Canine Mammary Carcinomas. Vet Pathol 2018; 55:622-633. [PMID: 29788797 DOI: 10.1177/0300985818776054] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is a dynamic process linked to metastasis in many tumor types, including mammary tumors. In this study, we evaluated E-cadherin and vimentin immunolocalization in primary canine mammary carcinomas (20 cases) and their respective metastases, as well as their relationship with the core regulators SNAIL/SLUG. To assess the number of cells undergoing the process of EMT, we quantitated double-positive (E-cadherin+/vimentin+) cells using immunofluorescence, via cell counting and image analysis. In addition, SNAIL/SLUG expression was evaluated by established immunohistochemical methods. Primary tumors had significantly more E-cadherin+/vimentin+ co-expression than their paired respective lymph node or distant metastasis, respectively. Furthermore, the percentage of E-cadherin+/vimentin+ cells in grade II and III carcinomas was significantly higher than in grade I tumors. Primary tumors had significantly higher SNAIL/SLUG expression when analyzed based on the percentage of positive cells compared with their respective distant metastases in pairwise comparisons. An inverse correlation was noted between SNAIL/SLUG immunoreactivity and percentage of E-cadherin+/vimentin+ immunopositive cells in primary tumor samples when SNAIL/SLUG immunoreactivity was grouped into 2 categories (high versus low) based on percentage-positive staining. These results show a positive correlation between E-cadherin+/vimentin+ cells and higher tumor grade, establish differences between primary tumor and their respective metastases, and provide further support that EMT plays a critical role in the metastasis of canine mammary carcinoma. Furthermore, these data suggest that modulation of this process could provide greater therapeutic control and provide support for further research to determine if E-cadherin+/vimentin+ co-immunoreactivity imparts predictive value in the clinical outcome of patients with canine mammary carcinomas.
Collapse
Affiliation(s)
- Talita M M Raposo-Ferreira
- 1 Department of Veterinary Clinic and Surgery, School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, Brazil
| | - Becky K Brisson
- 2 Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy C Durham
- 3 Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.,4 Penn Vet Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Renee Laufer-Amorim
- 5 Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, Brazil
| | - Veronica Kristiansen
- 6 Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Ellen Puré
- 4 Penn Vet Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.,7 Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan W Volk
- 2 Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.,4 Penn Vet Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.,7 Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karin Sorenmo
- 4 Penn Vet Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.,7 Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
48
|
Bednarczyk RB, Tuli NY, Hanly EK, Rahoma GB, Maniyar R, Mittelman A, Geliebter J, Tiwari RK. Macrophage inflammatory factors promote epithelial-mesenchymal transition in breast cancer. Oncotarget 2018; 9:24272-24282. [PMID: 29849939 PMCID: PMC5966261 DOI: 10.18632/oncotarget.24917] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 09/05/2017] [Indexed: 01/08/2023] Open
Abstract
The majority of breast cancers (90-95%) arise due to mediators distinct from inherited genetic mutations. One major mediator of breast cancer involves chronic inflammation. M1 macrophages are an integral component of chronic inflammation and the breast cancer tumor microenvironment (TME). Previous studies have demonstrated that up to 50% of the breast tumor comprise of tumor-associated macrophages (TAMs) and increased TAM infiltration has been associated with poor patient prognosis. Furthermore, breast cancer associated deaths are predominantly attributed to invasive cancers and metastasis with epithelial-mesenchymal transition (EMT) being implicated. In this study, we investigated the effects of cellular crosstalk between TAMs and breast cancer using an in vitro model system. M1 polarized THP-1 macrophage conditioned media (CM) was generated and used to evaluate cellular and functional changes of breast cancer lines T47D and MCF-7. We observed that T47D and MCF-7 exhibited a partial EMT phenotype in the presence of activated THP-1 CM. Additionally, MCF-7 displayed a significant increase in migratory and invasive properties. We conclude that M1 secretory factors can promote a partial EMT of epithelial-like breast cancer cells. The targeting of M1 macrophages or their secretory components may inhibit EMT and limit the invasive potential of breast cancer.
Collapse
Affiliation(s)
- Robert B Bednarczyk
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Neha Y Tuli
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Elyse K Hanly
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Ghada Ben Rahoma
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Rachana Maniyar
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Abraham Mittelman
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Jan Geliebter
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Raj K Tiwari
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
49
|
Gravel SP. Deciphering the Dichotomous Effects of PGC-1α on Tumorigenesis and Metastasis. Front Oncol 2018; 8:75. [PMID: 29629336 PMCID: PMC5876244 DOI: 10.3389/fonc.2018.00075] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/06/2018] [Indexed: 12/31/2022] Open
Abstract
Metabolic reprogramming confers cancer cells the ability to grow and survive under nutrient-depleted or stressful microenvironments. The amplification of oncogenes, the loss of tumor suppressors, as well as context- and lineage-specific determinants can converge and profoundly affect the metabolic status of cancer cells. Cumulating evidences suggest that highly glycolytic cells under the influence of oncogenes such as BRAF, or evolving in hypoxic microenvironments, will promote metastasis through modulation of multiple steps of tumorigenesis such as the epithelial-to-mesenchymal transition (EMT). On the contrary, increased reliance on mitochondrial respiration is associated with hyperplasic rather than metastatic disease. The PGC-1α transcriptional coactivator, a master regulator of mitochondrial biogenesis, has recently been shown to exert antimetastatic effects in cancer, notably through inhibition of EMT. Besides, PGC-1α has the opposite role in specific cancer subtypes, in which it appears to provide growth advantages. Thus, the regulation and role of PGC-1α in cancer is not univocal, and its use as a prognostic marker appears limited given its highly dynamic nature and its multifaceted regulation by transcriptional and posttranslational mechanisms. Herein, we expose key oncogenic and lineage-specific modules that finely regulate PGC-1α to promote or dampen the metastatic process. We propose a unifying model based on the systematic analysis of its controversial implication in cancer from cell proliferation to EMT and metastasis. This short review will provide a good understanding of current challenges associated with the study of PGC-1α.
Collapse
Affiliation(s)
- Simon-Pierre Gravel
- Laboratory of Metabolic Immunopharmacology, Faculty of Pharmacy, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
50
|
Sun B, Zhang D, Zhao N, Zhao X. Epithelial-to-endothelial transition and cancer stem cells: two cornerstones of vasculogenic mimicry in malignant tumors. Oncotarget 2018; 8:30502-30510. [PMID: 27034014 PMCID: PMC5444760 DOI: 10.18632/oncotarget.8461] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/14/2016] [Indexed: 01/11/2023] Open
Abstract
Vasculogenic mimicry (VM) is a functional microcirculation pattern in malignant tumors accompanied by endothelium-dependent vessels and mosaic vessels. VM has been identified in more than 15 solid tumor types and is associated with poor differentiation, late clinical stage and poor prognosis. Classic anti-angiogenic agents do not target endothelium-dependent vessels and are not efficacious against tumors exhibiting VM. Further insight into the molecular signaling that triggers and promotes VM formation could improve anti-angiogenic therapeutics. Recent studies have shown that cancer stem cells (CSCs) and epithelium-to-endothelium transition (EET), a subtype of epithelial-to-mesenchymal transition (EMT), accelerate VM formation by stimulating tumor cell plasticity, remodeling the extracellular matrix (ECM) and connecting VM channels with host blood vessels. VM channel-lining cells originate from CSCs due to expression of EMT inducers such as Twist1, which promote EET and ECM remodeling. Hypoxia and high interstitial fluid pressure in the tumor microenvironment induce a specific type of cell death, linearly patterned programmed cell necrosis (LPPCN), which spatially guides VM and endothelium-dependent vessel networks. This review focuses on the roles of CSCs and EET in VM, and on possible novel anti-angiogenic strategies against alternative tumor vascularization.
Collapse
Affiliation(s)
- Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|