1
|
Li H, Wang S, Li X, Weng Y, Guo D, Kong P, Cheng C, Wang Y, Zhang L, Cheng X, Cui Y. CDCA7 promotes TGF-β-induced epithelial-mesenchymal transition via transcriptionally regulating Smad4/Smad7 in ESCC. Cancer Sci 2022; 114:91-104. [PMID: 36056599 PMCID: PMC9807500 DOI: 10.1111/cas.15560] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 01/07/2023] Open
Abstract
Cell division cycle associated 7 (CDCA7) is a copy number amplification gene that contributes to the metastasis and invasion of tumors, including esophageal squamous cell carcinoma (ESCC). This present study aimed at clarifying whether high expression of CDCA7 promotes the metastasis and invasion of ESCC cell lines and exploring the underlying mechanisms implicated in epithelial-mesenchymal transition (EMT) of ESCC. The role of CDCA7 in the regulation of ESCC metastasis and invasion was evaluated using ESCC cell lines. Expression of EMT-related markers including E-cadherin, N-cadherin, Vimentin, Snail, and Slug, transforming growth factor β (TGF-β) signaling pathway including Smad2/3, p-Smad2/3, Smad4, and Smad7 were detected in CDCA7 knockdown and overexpressed cell lines. Dual-luciferase reporter assay and rescue assay were used to explore the underlying mechanisms that CDCA7 contributed to the metastasis and invasion of ESCC. High CDCA7 expression significantly promoted the metastasis and invasion of ESCC cell lines both in vivo and in vitro. Additionally, the expression of CDCA7 positively correlated with the expression of N-cadherin, Vimentin, Snail, Slug, TGF-β signaling pathway and negatively correlated with the expression of E-cadherin. Furthermore, CDCA7 transcriptionally regulated the expression of Smad4 and Smad7. Knockdown of CDCA7 inhibited the TGF-β signaling pathway and therefore inhibited EMT. Our data indicated that CDCA7 was heavily involved in EMT by regulating the expression of Smad4 and Smad7 in TGF-β signaling pathway. CDCA7 might be a new therapeutic target in the suppression of metastasis and invasion of ESCC.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Shaojie Wang
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Xiubo Li
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Yongjia Weng
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Dinghe Guo
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Pengzhou Kong
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Caixia Cheng
- Department of Pathology, The First HospitalShanxi Medical UniversityTaiyuanChina
| | - Yanqiang Wang
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Ling Zhang
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Xiaolong Cheng
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| | - Yongping Cui
- Department of Pathology, School of Basic Medical ScienceShanxi Medical UniversityTaiyuanChina,Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
| |
Collapse
|
2
|
Hu Y, Li Q, Yi K, Yang C, Lei Q, Wang G, Wang Q, Xu X. HuR Affects the Radiosensitivity of Esophageal Cancer by Regulating the EMT-Related Protein Snail. Front Oncol 2022; 12:883444. [PMID: 35664798 PMCID: PMC9160430 DOI: 10.3389/fonc.2022.883444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/21/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose We previously found that Hu antigen R (HuR) can regulate the proliferation and metastasis of esophageal cancer cells. This study aims to explore the effects of HuR on the radiosensitivity of esophageal cancer. Materials and Method Analyses of CCK-8, colony formation assay, Western blot, immunofluorescence, flow cytometry, reactive oxygen species (ROS), and mitochondrial membrane potential were conducted to characterize the esophageal cancer cells. Nude mouse models were used to detect the effects of HuR in a combination of X-ray treatment on the subcutaneous xenografts of esophageal cancer. In addition, a luciferase assay was used to detect the direct interaction of HuR with Snail mRNA 3’-UTR. Results The down-regulation of HuR combined with X-ray can significantly inhibit the proliferation and colony formation of esophageal cancer cells. Flow cytometry data showed that the down-regulation of HuR could induce a G1 phase cell cycle block in esophageal cancer cells, and aggravate X-ray-induced apoptosis, indicated by the increases of apoptosis-related proteins Bax, caspase-3 and caspase-9. Moreover, the down-regulation of HuR could significantly impair the mitochondrial membrane potential and increase the ROS production and DNA double-strand break marker γH2AX expression in esophageal cancer cells that were exposed to X-rays. In vivo data showed that the down-regulation of HuR combined with radiation significantly decreased the growth of subcutaneous xenograft tumors. Furthermore, HuR could interact with Snail. Up-regulation of Snail can reverse the EMT inhibitory effects caused by HuR down-regulation, and attenuate the tumor-inhibiting and radiosensitizing effects caused by HuR down-regulation. Conclusion In summary, our data demonstrate that HuR effectively regulates the radiosensitivity of esophageal cancer, which may be achieved by stabilizing Snail. Thus, HuR/Snail axis is a potentially therapeutic target for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Yan Hu
- Central Laboratory, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, China
| | - Qing Li
- Department of Gastroenterology, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, China
| | - Ke Yi
- Central Laboratory, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, China
| | - Chi Yang
- Department of General Surgery, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, China
| | - Qingjun Lei
- Department of General Surgery, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, China
| | - Guanghui Wang
- School of Pharmacy, Soochow University, Suzhou, China
| | - Qianyun Wang
- Department of Thoracic Surgery, the Third Affiliated Hospital to Soochow University, Changzhou, China
| | - Xiaohui Xu
- Central Laboratory, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, China.,Department of General Surgery, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, China
| |
Collapse
|
3
|
Kadian LK, Arora M, Prasad CP, Pramanik R, Chauhan SS. Signaling pathways and their potential therapeutic utility in esophageal squamous cell carcinoma. Clin Transl Oncol 2022; 24:1014-1032. [PMID: 34990001 DOI: 10.1007/s12094-021-02763-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022]
Abstract
Esophageal cancer is a complex gastrointestinal malignancy with an extremely poor outcome. Approximately 80% of cases of this malignancy in Asian countries including India are of squamous cell origin, termed Esophageal Squamous Cell Carcinoma (ESCC).The five-year survival rate in ESCC patients is less than 20%. Neo-adjuvant chemo-radiotherapy (NACRT) followed by surgical resection remains the major therapeutic strategy for patients with operable ESCC. However, resistance to NACRT and local recurrence after initial treatment are the leading cause of dismal outcomes in these patients. Therefore, an alternative strategy to promote response to the therapy and reduce the post-operative disease recurrence is highly needed. At the molecular level, wide variations have been observed in tumor characteristics among different populations, nevertheless, several common molecular features have been identified which orchestrate disease progression and clinical outcome in the malignancy. Therefore, determination of candidate molecular pathways for targeted therapy remains the mainstream idea of focus in ESCC research. In this review, we have discussed the key signaling pathways associated with ESCC, i.e., Notch, Wnt, and Nrf2 pathways, and their crosstalk during disease progression. We further discuss the recent developments of novel agents to target these pathways in the context of targeted cancer therapy. In-depth research of the signaling pathways, gene signatures, and a combinatorial approach may help in discovering targeted therapy for ESCC.
Collapse
Affiliation(s)
- L K Kadian
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - M Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - C P Prasad
- Department of Medical Oncology (Lab), Dr. B. R. Ambedkar-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - R Pramanik
- Department of Medical Oncology, Dr. B. R. Ambedkar-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - S S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
4
|
Zada S, Hwang JS, Lai TH, Pham TM, Ahmed M, Elashkar O, Kim W, Kim DR. Autophagy-mediated degradation of NOTCH1 intracellular domain controls the epithelial to mesenchymal transition and cancer metastasis. Cell Biosci 2022; 12:17. [PMID: 35164848 PMCID: PMC8842742 DOI: 10.1186/s13578-022-00752-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
Backgound Autophagy controls levels of cellular components during normal and stress conditions; thus, it is a pivotal process for the maintenance of cell homeostasis. In cancer, autophagy protects cells from cancerous transformations that can result from genomic instability induced by reactive oxygen species or other damaged components, but it can also promote cancer survival by providing essential nutrients during the metabolic stress condition of cancer progression. However, the molecular mechanism underlying autophagy-dependent regulation of the epithelial to mesenchymal transition (EMT) and metastasis is still elusive. Methods The intracellular level of NOTCH1 intracellular domain (NICD) in several cancer cells was studied under starvation, treatment with chloroquine or ATG7-knockdown. The autophagy activity in these cells was assessed by immunocytochemistry and molecular analyses. Cancer cell migration and invasion under modulation of autophagy were determined by in vitro scratch and Matrigel assays. Results In the study, autophagy activation stimulated degradation of NICD, a key transcriptional regulator of the EMT and cancer metastasis. We also found that NICD binds directly to LC3 and that the NICD/LC3 complex associates with SNAI1 and sequestosome 1 (SQSTM1)/p62 proteins. Furthermore, the ATG7 knockdown significantly inhibited degradation of NICD under starvation independent of SQSTM1-associated proteasomal degradation. In addition, NICD degradation by autophagy associated with the cellular level of SNAI1. Indeed, autophagy inhibited nuclear translocation of NICD protein and consequently decreased the transcriptional activity of its target genes. Autophagy activation substantially suppressed in vitro cancer cell migration and invasion. We also observed that NICD and SNAI1 levels in tissues from human cervical and lung cancer patients correlated inversely with expression of autophagy-related proteins. Conclusions These findings suggest that the cellular level of NICD is regulated by autophagy during cancer progression and that targeting autophagy-dependent NICD/SNAI1 degradation could be a strategy for the development of cancer therapeutics. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00752-3.
Collapse
Affiliation(s)
- Sahib Zada
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea.,Cancer Biology and Immunology Laboratory, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Trang Minh Pham
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Omar Elashkar
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Wanil Kim
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea.
| |
Collapse
|
5
|
Gao K, Xing W, Liu X, Liu J, Sun H, Hao W, Zheng Y. The Notch1 gene may control cell chemoresistance in esophageal squamous cell cancer. Transl Cancer Res 2021; 10:3278-3285. [PMID: 35116634 PMCID: PMC8799163 DOI: 10.21037/tcr-21-447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/04/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND In our previous study, missense mutations in the Notch1 gene were found in chemotherapy-resistant esophageal squamous cell cancer (ESCC) patients. In this study, we explored changes in the interaction between Notch1 and DLL4 resulting from missense mutations. METHODS Bioinformatics analysis was performed to assess and compare the different biological structures and functions of wild type (WT) and mutation type (MT) sequences of Notch1. A genetic information search was performed, and the results were analyzed using in silico modeling. Homology modeling of the Notch1 protein was carried out using Swiss-Model software, and modeling of site-directed mutations was carried out using PyMOL software to observe the protein structure. The Notch1-DLL4 ligand-receptor complex protein model was constructed, Wincoot software was used to determine site-directed mutations, and a protein-ligand interaction profiler (PLIP) was used to calculate the noncovalent interactions in the complex. RESULTS The mutation site was located in the region where Notch1 binds to DLL4. A careful examination of the in silico structural model revealed that the mutation caused an alteration in the surface charge, and the water-bridge bonds of the interaction between Notch1-DLL4 increased in number from 5 to 7. CONCLUSIONS Notch1 gene missense mutation leads to an increase in the number of water-bridge bonds, thus enhancing the Notch1-DLL4 interaction, which may lead to tighter Notch1-DLL4 binding, either making the pathway easier to activate or increasing the length of time it is active.
Collapse
Affiliation(s)
- Kun Gao
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenqun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xianben Liu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jizhao Liu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Haibo Sun
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wentao Hao
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yan Zheng
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
6
|
Li X, Wang M, Li S, Chen Y, Wang M, Wu Z, Sun X, Yao L, Dong H, Song Y, Xu Y. HIF-1-induced mitochondrial ribosome protein L52: a mechanism for breast cancer cellular adaptation and metastatic initiation in response to hypoxia. Am J Cancer Res 2021; 11:7337-7359. [PMID: 34158854 PMCID: PMC8210597 DOI: 10.7150/thno.57804] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/16/2021] [Indexed: 01/16/2023] Open
Abstract
Background: Hypoxia is a hallmark of the physical microenvironment of solid tumors. As a key factor that regulates tumor development and progression, hypoxia can reprogram the expression of multiple genes, whose biological function and molecular mechanism in cancer remain largely unclear. The mitochondrial ribosome protein family consists of nuclear-encoded mitochondrial proteins that are responsible for protein synthesis in the mitochondria. Methods: A high-throughput RNA sequencing assay was carried out to identify differentially expressed mRNAs between breast cancer tissues and adjacent normal tissues as well as breast tumors with metastasis and those without metastasis. Our clinical samples and TCGA database were analyzed to observe the clinical value of mitochondrial ribosome protein L52 (MRPL52) in human breast cancer. Potent hypoxia response elements in the promoter region of MRPL52 were identified and validated by chromatin immunoprecipitation and luciferase reporter assays. Functional experiments were performed using breast cancer cell lines with MRPL52 ectopic expression and knockdown cultured in a 20% or 1% O2 environment. Results: MRPL52 expression was upregulated in human breast cancer and was significantly associated with aggressive clinicopathological characteristics and a higher metastatic risk of breast cancer patients. We found that the overexpression of MRPL52 in breast cancer is induced by hypoxia-inducible factor-1 in response to hypoxic exposure. The role of MRPL52 in suppressing apoptosis and promoting migration and invasion of hypoxic breast cancer cells was demonstrated by our experimental evidence. Mechanistically, MRPL52 promoted PTEN-induced putative kinase 1 /Parkin-dependent mitophagy to remove oxidatively damaged mitochondria and prevent uncontrolled reactive oxygen species (ROS) generation, thus repressing activation of the mitochondrial apoptotic cascade. Additionally, MRPL52 augmented epithelial-mesenchymal transition, migration and invasion of hypoxic breast cancer cells by activating the ROS-Notch1-Snail signaling pathway. Benefited from this bidirectional regulatory mechanism, MRPL52 is responsible for maintaining ROS levels in a window that can induce tumorigenic signal transduction without causing cytotoxicity in hypoxic breast cancer cells. Conclusions: This work elucidates the molecular mechanism by which MRPL52 mediates hypoxia-induced apoptotic resistance and metastatic initiation of breast cancer, and provides new insights into the interplay between cancer and the tumor microenvironment.
Collapse
|
7
|
Lu YB, Sun TJ, Chen YT, Cai ZY, Zhao JY, Miao F, Yang YN, Wang SX. Targeting the Epithelial-to-Mesenchymal Transition in Cancer Stem Cells for a Better Clinical Outcome of Glioma. Technol Cancer Res Treat 2020; 19:1533033820948053. [PMID: 33089751 PMCID: PMC7586027 DOI: 10.1177/1533033820948053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/19/2020] [Accepted: 07/09/2020] [Indexed: 02/05/2023] Open
Abstract
Glioma is one of the most common malignant tumors of the central nervous system with a poor prognosis at present due to lack of effective treatment options. Its initiation, migration, and multipotency are affected by cancer stem cell's transition. Previous studies imply that changes in the cancer stem cells can affect the malignant differentiation of the tumor. We found that the epithelial-to-mesenchymal transition (EMT)-related regulatory pathway is an important target for tumor therapy. In this review, we discuss the transition factor of EMT and 3 specific pathways that affect the EMT of cancer stem cells during tumor development. We conclude that targeting the EMT process of cancer stem cells can be a feasible approach in the treatment of glioma.
Collapse
Affiliation(s)
- Yu-bao Lu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- *Both authors contributed equally to this study and share first authorship
| | - Tian-Jiao Sun
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- *Both authors contributed equally to this study and share first authorship
| | - Yu-tong Chen
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Zong-Yan Cai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jia-Yu Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Feng Miao
- Zhangye People's Hospital Affiliated to Hexi University, Zhangye, Gansu, China
| | - Yong-na Yang
- Department of Neurology, The First People’s Hospital of Lanzhou City, Lanzhou, Gansu, China
| | - Shi-Xin Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
8
|
Retraction Note: RNAi targeting CXCR4 inhibits proliferation and invasion of esophageal carcinoma cells. Diagn Pathol 2019; 14:44. [PMID: 31109345 PMCID: PMC6528241 DOI: 10.1186/s13000-019-0828-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
9
|
Jing L, Ruan Z, Sun H, Li Q, Han L, Huang L, Yu S, Wang Y, Guo H, Jiao M. Epithelial-mesenchymal transition induced cancer-stem-cell-like characteristics in hepatocellular carcinoma. J Cell Physiol 2019; 234:18448-18458. [PMID: 30908631 DOI: 10.1002/jcp.28480] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma in China accounts for half of the world's incidence. Both epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs) are thought to be involved in tumor malignant progression. However, the relationship between EMT and CSCs is still unclear. Bioinformatics analysis was performed to evaluate the relationship between EMT and CSCs. The EMT and CSC regulatory mechanism was investigated through Transwell, wound-healing, sphere formation, colony-forming, and western blotting assays. Immunofluorescence and immunoprecipitation were used to study the interaction of hypoxia inducible factor 1α (HIF-1α) /Notch1. Immunohistochemical study was applied to investigate the expression pattern in the process of hepatocellular carcinogenesis and development. In our present study, bioinformatics results indicate that the expression of EMT-related molecules is correlated with CSCs. In vitro studies indicated that EMT activation could induce CSC characteristics. Notch1 was confirmed to mediate the process of EMT-induced CSCs through the interaction with HIF-1α directly. Our findings indicate that EMT could induce CSC-like characteristics, which is mediated by HIF-1α-upregulated Notch intracellular domain expression.
Collapse
Affiliation(s)
- Li Jing
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhiping Ruan
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haifeng Sun
- The Third Department of Medical Oncology, Shaanxi Provincial Tumor Hospital, Xi'an, China
| | - Qing Li
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lili Han
- Department of Oncology, he Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lanxuan Huang
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Sizhe Yu
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yu Wang
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hui Guo
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Min Jiao
- Department of Medical Oncology, he First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Common profiles of Notch signaling differentiate disease-free survival in luminal type A and triple negative breast cancer. Oncotarget 2018; 8:6013-6032. [PMID: 27888801 PMCID: PMC5351609 DOI: 10.18632/oncotarget.13451] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/29/2016] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BC) is characterized by high heterogeneity regarding its biology and clinical characteristics. The Notch pathway regulates such processes as organ modeling and epithelial-to-mesenchymal transition (EMT). The aim of the study was to determine the effect of differential expression of Notch members on disease-free survival (DFS) in luminal type A (lumA) and triple negative (TN) BC. The differential expression of 19 Notch members was examined in a TCGA BC cohort. DFS analysis was performed using the log-rank test (p<0.05). Biological differences between DFS groups were determined with Gene Set Enrichment Analysis (GSEA) (tTest, FDR<0.25). Common expression profiles according to Notch signaling were examined using ExpressCluster (K-means, mean centered, Euclidean distance metric). The overexpression of HES1, LFNG and PSEN1 was found to be favorable for DFS in lumA, and lowered expression favorable for DFS in TN. GSEA analysis showed that differential Notch signaling is associated with cell cycle, tissue architecture and remodeling. Particularly, targets of E2F, early stage S phase transcription factor, were upregulated in the lumA unfavorable group and the TN favorable group differentiated on a basis of HES1 and PSEN1 expression. Summarizing, our analysis show significance of Notch signaling in BRCA progression through triggering EMT. Moreover, identification of numerous genes which overexpression is associated with disease recurrence may serve as a source of potential targets for a new anticancer therapy.
Collapse
|
11
|
Wang L, Hou Z, Hasim A, Abuduerheman A, Zhang H, Niyaz M, Awut I, Upur H, Sheyhidin I. RNF113A promotes the proliferation, migration and invasion, and is associated with a poor prognosis of esophageal squamous cell carcinoma. Int J Oncol 2018; 52:861-871. [PMID: 29393393 DOI: 10.3892/ijo.2018.4253] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 12/04/2017] [Indexed: 11/06/2022] Open
Abstract
Ring finger protein 113A (RNF113A) possesses a C3HC4 zinc finger domain and this domain is found in E3 ubiquitin ligase and is involved in tumorigenesis. To date, and at least to the best of our knowledge, there are no studies available which have investigated RNF113A in cancer. Thus, this study aimed to explore the role of RNF113A in the development of esophageal squamous cell carcinoma (ESCC). For this purpose, paraffin-embedded samples from 117 patients with ESCC were selected, as well as 41 pairs of fresh-frozen ESCC and adjacent normal tissue samples. RNF113A expression was examined by immunohistochemistry and reverse transcription-quantitative PCR (RT-qPCR). RNF113A was overexpressed or silenced in the EC9706 and Eca109 cells. The cells were examined for cell cycle progression, apoptosis, invasiveness and migration. Xenograft tumors were also created in mice using the Eca109 cells. Tumor differentiation (P=0.008) and T classification (P<0.001) were found to be significantly associated with RNF113A expression. No statistically significant association was observed between RNF113A expression and sex, age, histological type, tumor location and lymph node metastasis (N classification). Kaplan-Meier analysis revealed that the patients with ESCC with ahigh expression of RNF113A had a lower survival rate than those with a low expression (P=0.002). Multivariate analysis revealed that RNF113A expression (HR=2.406; 95% CI, 1.301-4.449, P=0.005) was independently associated with overall survival in patients with ESCC. The overexpression of RNF113A promoted proliferation, migration, and invasiveness of ESCC cell lines in vitro, and RNF113A silencing reversed these malignant behaviors. RNF113A knockdown inhibited tumor growth in vivo. Thus, these results indicate that RNF113A promotes the proliferation, migration and invasiveness of ESCC cell lines. RNF113A expression in ESCC is this associated with a poor prognosis of affected patients.
Collapse
Affiliation(s)
- Lei Wang
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Zhichao Hou
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Ayshamgul Hasim
- Department of Pathology, Medical University of Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Abulajiang Abuduerheman
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Haiping Zhang
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Madiniyat Niyaz
- Clinical Medical Research Institute, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Idiris Awut
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Halmurat Upur
- Department of Uyghur Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Ilyar Sheyhidin
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| |
Collapse
|
12
|
Natsuizaka M, Whelan KA, Kagawa S, Tanaka K, Giroux V, Chandramouleeswaran PM, Long A, Sahu V, Darling DS, Que J, Yang Y, Katz JP, Wileyto EP, Basu D, Kita Y, Natsugoe S, Naganuma S, Klein-Szanto AJ, Diehl JA, Bass AJ, Wong KK, Rustgi AK, Nakagawa H. Interplay between Notch1 and Notch3 promotes EMT and tumor initiation in squamous cell carcinoma. Nat Commun 2017; 8:1758. [PMID: 29170450 PMCID: PMC5700926 DOI: 10.1038/s41467-017-01500-9] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/21/2017] [Indexed: 12/15/2022] Open
Abstract
Notch1 transactivates Notch3 to drive terminal differentiation in stratified squamous epithelia. Notch1 and other Notch receptor paralogs cooperate to act as a tumor suppressor in squamous cell carcinomas (SCCs). However, Notch1 can be stochastically activated to promote carcinogenesis in murine models of SCC. Activated form of Notch1 promotes xenograft tumor growth when expressed ectopically. Here, we demonstrate that Notch1 activation and epithelial–mesenchymal transition (EMT) are coupled to promote SCC tumor initiation in concert with transforming growth factor (TGF)-β present in the tumor microenvironment. We find that TGFβ activates the transcription factor ZEB1 to repress Notch3, thereby limiting terminal differentiation. Concurrently, TGFβ drives Notch1-mediated EMT to generate tumor initiating cells characterized by high CD44 expression. Moreover, Notch1 is activated in a small subset of SCC cells at the invasive tumor front and predicts for poor prognosis of esophageal SCC, shedding light upon the tumor promoting oncogenic aspect of Notch1 in SCC. Notch receptors can exert different roles in cancer. In this manuscript, the authors reveal that Notch1 activation and EMT promote tumor initiation and cancer cell heterogeneity in squamous cell carcinoma, while the repression of Notch3 by ZEB1 limits Notch1-induced differentiation, permitting Notch1-mediated EMT.
Collapse
Affiliation(s)
- Mitsuteru Natsuizaka
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, 060-8638, Japan
| | - Kelly A Whelan
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Shingo Kagawa
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Chiba, 260-0856, Japan
| | - Koji Tanaka
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Department of Surgery, Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Veronique Giroux
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Prasanna M Chandramouleeswaran
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Apple Long
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Varun Sahu
- Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Douglas S Darling
- Department of Oral Immunology and Infectious Diseases, and Center for Genetics and Molecular Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Jianwen Que
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University, New York, NY, 10032, USA
| | - Yizeng Yang
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jonathan P Katz
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - E Paul Wileyto
- Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Devraj Basu
- Abramson Cancer Center, Philadelphia, PA, 19104, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yoshiaki Kita
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8520, Japan
| | - Shoji Natsugoe
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8520, Japan
| | - Seiji Naganuma
- Department of Pathology, Kochi Medical School, Nankoku-shi, Kochi, 783-8505, Japan
| | - Andres J Klein-Szanto
- Histopathology Facility and Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - J Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Adam J Bass
- Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Kwok-Kin Wong
- Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA. .,Division of Hematology and Medical Oncology, New York University, New York, NY, 10016, USA.
| | - Anil K Rustgi
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Hiroshi Nakagawa
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
13
|
Lubin DJ, Mick R, Shroff SG, Stashek K, Furth EE. The notch pathway is activated in neoplastic progression in esophageal squamous cell carcinoma. Hum Pathol 2017; 72:66-70. [PMID: 29137934 DOI: 10.1016/j.humpath.2017.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/16/2022]
Abstract
The Notch signaling pathway is integral to normal human development and homeostasis and has a deterministic function on cell differentiation. Recent studies suggest aberrant Notch signaling may contribute to neoplastic progression by an increase in stem cell survival, chemoresistance, and the promotion of epithelial-to-mesenchymal transition. The goals of our study were to determine, utilizing quantitative technologies, the expression of activated Notch 1 in esophageal squamous cell carcinoma (SCC) and to determine the relationship between Notch 1 expression and various clinicopathologic parameters. Immunohistochemical staining for Notch intracellular domain (NICD) was performed on 60 consecutive cases of esophageal squamous cell carcinoma, 42 cases of benign esophageal squamous epithelium, and 13 cases of eosinophilic esophagitis diagnosed in our department from 2007 through 2015, and exact nuclear staining and nuclear characteristics were graded using the Vectra imaging system. Clinicopathologic data (gender, age at diagnosis, smoking status, tumor grade, tumor stage, tumor location, and survival) were collected for each SCC case and these were correlated with NICD staining. Cases of esophageal SCC demonstrated significantly higher NICD staining compared to cases of benign and reactive esophageal epithelium (P=.003 and .005, respectively). Among cases of esophageal SCC, nuclear NICD staining was significantly correlated with both tumor grade and stage. Following classification and regression tree analysis, esophageal SCC patients with increased NICD expression were found to be more likely to die from their disease than those with lower levels of expression. Taken together, the findings suggest that increased Notch 1 may contribute to the development and aggressiveness of esophageal SCC.
Collapse
Affiliation(s)
- Daniel J Lubin
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104.
| | - Rosemarie Mick
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, 19104
| | - Stuti G Shroff
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104
| | - Kristen Stashek
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104
| | - Emma E Furth
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
14
|
Huang T, Zhou Y, Cheng ASL, Yu J, To KF, Kang W. NOTCH receptors in gastric and other gastrointestinal cancers: oncogenes or tumor suppressors? Mol Cancer 2016; 15:80. [PMID: 27938406 PMCID: PMC5148895 DOI: 10.1186/s12943-016-0566-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/01/2016] [Indexed: 12/18/2022] Open
Abstract
Gastric cancer (GC) ranks the most common cancer types and is one of the leading causes of cancer-related death. Due to delayed diagnosis and high metastatic frequency, 5-year survival rate of GC is rather low. It is a complex disease resulting from the interaction between environmental factors and host genetic alterations that deregulate multiple signaling pathways. The Notch signaling pathway, a highly conserved system in the regulation of the fate in several cell types, plays a pivotal role in cell differentiation, survival and proliferation. Notch is also one of the most commonly activated signaling pathways in tumors and its aberrant activation plays a key role in cancer advancement. Whether Notch cascade exerts oncogenic or tumor suppressive function in different cancer types depends on the cellular context. Mammals have four NOTCH receptors that modulate Notch pathway activity. In this review, we provide a comprehensive summary on the functional role of NOTCH receptors in gastric and other gastrointestinal cancers. Increasing knowledge of NOTCH receptors in gastrointestinal cancers will help us recognize the underlying mechanisms of Notch signaling and develop novel therapeutic strategies for GC.
Collapse
Affiliation(s)
- Tingting Huang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, SAR, People's Republic of China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Yuhang Zhou
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, SAR, People's Republic of China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - Alfred S L Cheng
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Jun Yu
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, SAR, People's Republic of China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, SAR, People's Republic of China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
| |
Collapse
|
15
|
Liu X, Yun F, Shi L, Li ZH, Luo NR, Jia YF. Roles of Signaling Pathways in the Epithelial-Mesenchymal Transition in Cancer. Asian Pac J Cancer Prev 2016; 16:6201-6. [PMID: 26434817 DOI: 10.7314/apjcp.2015.16.15.6201] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a cellular process though which an epithelial phenotype can be converted into a phenotype of mesenchymal cells. Under physiological conditions EMT is important for embryogenesis, organ development, wound repair and tissue remodeling. However, EMT may also be activated under pathologic conditions, especially in carcinogenesis and metastatic progression. Major signaling pathways involved in EMT include transforming growth factor β(TGF-β), Wnt, Notch, Hedgehog and other signaling pathways. These pathways are related to several transcription factors, including Twist, Smads and zinc finger proteins snail and slug. These interact with each other to provide crosstalk between the relevant signaling pathways. This review lays emphasis on studying the relationship between EMT and signaling pathways in carcinogenesis and metastatic progression.
Collapse
Affiliation(s)
- Xia Liu
- Department of Pathology, The First Affiliated Hospital of Inner Mongolia Medical University, Huhhot, China E-mail :
| | | | | | | | | | | |
Collapse
|
16
|
Qi J, Li T, Bian H, Li F, Ju Y, Gao S, Su J, Ren W, Qin C. SNAI1 promotes the development of HCC through the enhancement of proliferation and inhibition of apoptosis. FEBS Open Bio 2016; 6:326-37. [PMID: 27239445 PMCID: PMC4821356 DOI: 10.1002/2211-5463.12043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 01/08/2023] Open
Abstract
SNAI1, a zinc‐finger transcription factor, plays an important role in the induction of epithelial–mesenchymal transition (EMT) in various cancers. However, the possible functions of SNAI1 in the proliferation and apoptosis of hepatocellular carcinoma have not been clearly identified. In this study, we investigated the effects and mechanisms of SNAI1 in the proliferation and apoptosis of hepatocellular carcinoma using clinical samples and cell lines. We found that SNAI1 is highly expressed in the tissues of liver cancer compared with adjacent nontumor tissues. SNAI1 is also highly expressed in the hepatoma cell lines HepG2, SMMC‐7721, and BEL‐7402 compared with the human normal liver cell line L02. We also observed that SNAI1 expression was correlated with distal metastasis, incomplete tumor capsule formation, and histological differentiation in hepatocellular carcinoma (HCC). Moreover, we demonstrated that knockdown of SNAI1 via lentiviral vectors of RNAi against SNAI inhibited cell proliferation by inducing G1 arrest, which was accompanied by the downregulation of cyclin D1 but not that of cyclin A. In addition, knockdown of SNAI1 promoted apoptosis by decreasing the expression of Bcl‐2. In conclusion, our findings revealed that SNAI1 is involved in the development of hepatocellular carcinoma via regulating the growth and apoptosis of tumor cells.
Collapse
Affiliation(s)
- Jianni Qi
- Central Laboratory Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Tao Li
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Hongjun Bian
- Department of Emergency Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Feifei Li
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Ying Ju
- Department of Clinical Laboratory Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Shanshan Gao
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Jingran Su
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Wanhua Ren
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| | - Chengyong Qin
- Department of Gastroenterology Shandong Provincial Hospital Affiliated to Shandong University Jinan Shandong China
| |
Collapse
|
17
|
Ma Y, Bian J, Zhang F. Inhibition of perillyl alcohol on cell invasion and migration depends on the Notch signaling pathway in hepatoma cells. Mol Cell Biochem 2015; 411:307-15. [PMID: 26475687 DOI: 10.1007/s11010-015-2593-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/08/2015] [Indexed: 12/12/2022]
Abstract
Cell metastasis, especially the process of invasion and migration, is considered as the main cause for the high mortality rate of hepatocellular carcinoma (HCC), which has become the sixth most common cancer worldwide and the third leading cause of cancer death. In this present study, we aimed to exploit the effects of perillyl alcohol on cell invasion and migration and the underlying molecular mechanisms in HCC. According to the transwell assays, cell invasiveness and migratory capacity were markedly higher in hepatoma cells (HepG2, SMMC-7721 and MHCC97H) than those in normal liver cells (HL-7702), and then significantly suppressed by perillyl alcohol treatment (P < 0.05). Meanwhile, the mRNA levels of Notch signaling pathway downstream target genes, HES1, HES5, and HEY1, were notably higher in hepatoma cells detected with real-time reverse transcription polymerase chain reaction (RT-PCR) (P < 0.05). After treated with perillyl alcohol, these mRNA levels were significantly decreased in hepatoma cells (P < 0.05). In addition, compared with the normal liver cells, the protein expression levels of Notch1 intracellular domain (N1ICD) and Snail were significantly increased, while E-cadherin protein expression was significantly decreased in hepatoma cells (P < 0.05). However, perillyl alcohol treatment significantly decreased N1ICD and Snail protein expressions and increased E-cadherin protein expression in hepatoma cells (P < 0.05). In conclusion, perillyl alcohol might play an important role in the process of hepatoma cell invasion and migration via decreasing the activity of Notch signaling pathway and increasing E-cadherin expression regulated by Snail.
Collapse
Affiliation(s)
- Yong Ma
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Jianmin Bian
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Feng Zhang
- Department of General Surgery, The First Affiliated Hospital, Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
18
|
Chen W, Cao G, Yuan X, Zhang X, Zhang Q, Zhu Y, Dong Z, Zhang S. Notch-1 knockdown suppresses proliferation, migration and metastasis of salivary adenoid cystic carcinoma cells. J Transl Med 2015; 13:167. [PMID: 25990317 PMCID: PMC4445799 DOI: 10.1186/s12967-015-0520-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 05/06/2015] [Indexed: 11/24/2022] Open
Abstract
Background Notch-1 promotes invasion and metastasis of cancer cells but its role in salivary adenoid cystic carcinoma (SACC) remains unelucidated. Here, we sought to investigate the effect of Notch-1 knockdown on the invasion and metastasis of SACC cells. Methods Stable ACC-M cells whose Notch-1 was silenced by lentiviral vectors were established. Cellular proliferation was evaluated by the MTT assays and clonogenic assays, apoptosis by flow cytometry and the migration of ACC-M cells by Transwell assays. Metastasis was evaluated by examining the number of lung nodules in Balb⁄c nu⁄nu nude mice bearing subcutaneous SACC xenografts. Results Our MTT assay revealed that Notch-1 knockdown significantly suppressed the proliferation of ACC-M cells compared with non-infected or scrambled control cells. Clonogenic assays further showed that Notch-1 knockdown significantly suppressed the clonogenic growth of ACC-M cells (p < 0.01 vs. controls). Our flow cytometry demonstrated that Notch-1 knockdown was associated with a significantly higher proportion of late apoptotic and necrotic cells (p < 0.01 vs. controls). Transwell assays revealed that Notch-1 knockdown markedly reduced the migratory capacity of ACC-M cells (p < 0.01 vs. controls) and xenograft studies showed that the number of metastatic nodules in the lung surface was significantly lower in nude mice bearing xenografts with Notch-1 knockdown compared to those bearing control xenografts (p < 0.01 vs. controls). Conclusion Notch-1 knockdown suppresses the growth and migration of SACC cells in vitro and the metastasis of SACC cells in vivo. Notch-1 may be a new candidate target in SACC.
Collapse
Affiliation(s)
- Wei Chen
- Department of Stomatology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Gang Cao
- Department of Stomatology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Xinran Yuan
- Department of Immunology & Rheumatology, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China.
| | - Xiang Zhang
- Department of Stomatology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Qingqing Zhang
- Department of Stomatology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Yinglan Zhu
- Department of Stomatology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Zhen Dong
- Department of Stomatology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Senlin Zhang
- Department of Stomatology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| |
Collapse
|
19
|
Shao S, Zhao X, Zhang X, Luo M, Zuo X, Huang S, Wang Y, Gu S, Zhao X. Notch1 signaling regulates the epithelial-mesenchymal transition and invasion of breast cancer in a Slug-dependent manner. Mol Cancer 2015; 14:28. [PMID: 25645291 PMCID: PMC4322803 DOI: 10.1186/s12943-015-0295-3] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/13/2015] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) is crucial for the invasion and metastasis of breast cancer. However, how Notch signaling regulates the EMT process and invasion in breast cancer remains largely unknown. METHODS The impact of Notch1 silencing by specific shRNAs on the EMT and invasion of human breast cancer MCF-7 and MDA-MB-231 cells as well as xenografts was tested by western blot, real-time polymerase chain reaction (RT-PCR), immunofluorescence, transwell, and immunohistochemistry assays. The effect of Slug silencing or upregulation on the EMT and invasion of breast cancer cells was analyzed, and the effect of Notch1 signaling on Slug expression was determined by the luciferase reporter assay. RESULTS The Notch1 intracellular domain (N1ICD) and Jagged1 were expressed in breast cancer cells. Notch1 silencing reversed the spontaneous EMT process and inhibited the migration and invasion of breast cancer cells and the growth of xenograft breast cancers. The expression of N1ICD was upregulated significantly by Jagged1-mediated Notch signaling activation. Moreover, Jagged1-mediated Notch signaling promoted the EMT process, migration, and invasion of breast cancer cells, which were abrogated by Notch silencing. Furthermore, the N1ICD positively regulated the Slug expression by inducing Slug promoter activation. Importantly, the knockdown of Slug weakened the invasion ability of breast cancer cells and reversed the Jagged1-induced EMT process with significantly decreased expression of vimentin and increased expression of E-cadherin. In addition, Slug overexpression restored the Notch1 knockdown-suppressed EMT process. CONCLUSIONS Our novel data indicate that Notch signaling positively regulates the EMT, invasion, and growth of breast cancer cells by inducing Slug expression. The Notch1-Slug signaling axis may represent a potential therapeutic target for breast cancer therapy.
Collapse
Affiliation(s)
- Shan Shao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaoai Zhao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaojin Zhang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Minna Luo
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaoxiao Zuo
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Shangke Huang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Ying Wang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Shanzhi Gu
- The Department of Forensic Medicine, Medical School, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xinhan Zhao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
20
|
Takamine K, Ueda Y, Nakano K, Ochiai T, Sugita Y, Kubo K, Maeda H, Hasegawa H, Kawakami T. Notch as a Possible Cell Differentiation Factor in Pleomorphic Adenomas. Int J Med Sci 2015; 12:759-63. [PMID: 26516303 PMCID: PMC4615235 DOI: 10.7150/ijms.12882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/09/2015] [Indexed: 02/04/2023] Open
Abstract
The expression of Notch in 30 cases of pleomorphic adenoma was examined by immunohistochemistry. Comparing the results of our study with previous literatures, from the partial CK7 expression and substantial Notch expression in ductal epithelial cells as well as the Notch expression in solid tumor nests, it can be inferred that Notch is involved in cell differentiation. CK13 expression was observed in cells undergoing squamous metaplasia and Notch expression was seen in the nucleus of basal and squamous cells. The intense Notch expression in basal cells and weak expression in squamous cells suggests that Notch is involved in the differentiation from basal to squamous cell. Moreover, the loss of nuclear expression on the inner layer would signify that differentiation is about to end or has been terminated. Notch was expressed in the cytoplasm of cartilage cells and in the cell membrane of mucous cells but not in the nucleus indicating that differentiation has been concluded. Notch involvement is suspected in cell differentiation in areas showing ductal structures and squamous metaplasia. In summary, Notch is involved in cell differentiation of ductal cells in PA. Nuclear expression was shown in tumor cells in solid nests and surrounding structures. Moreover, Notch is expressed by basal cells undergoing squamous metaplasia suggesting the participation of Notch in cell differentiation in PA.
Collapse
Affiliation(s)
- Keisuke Takamine
- 1. Hard Tissue Pathology Unit, Matsumoto Dental University Graduate School of Oral Medicine, Shiojiri, Japan
| | - Yukiko Ueda
- 1. Hard Tissue Pathology Unit, Matsumoto Dental University Graduate School of Oral Medicine, Shiojiri, Japan
| | - Keisuke Nakano
- 1. Hard Tissue Pathology Unit, Matsumoto Dental University Graduate School of Oral Medicine, Shiojiri, Japan ; 2. Department of Oral Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmacuitical Sciences, Okayama, Japan
| | - Takanaga Ochiai
- 1. Hard Tissue Pathology Unit, Matsumoto Dental University Graduate School of Oral Medicine, Shiojiri, Japan
| | - Yoshihiko Sugita
- 3. Department of Oral Pathology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Katsutoshi Kubo
- 3. Department of Oral Pathology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Hatsuhiko Maeda
- 3. Department of Oral Pathology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Hiromasa Hasegawa
- 1. Hard Tissue Pathology Unit, Matsumoto Dental University Graduate School of Oral Medicine, Shiojiri, Japan
| | - Toshiyuki Kawakami
- 1. Hard Tissue Pathology Unit, Matsumoto Dental University Graduate School of Oral Medicine, Shiojiri, Japan
| |
Collapse
|
21
|
Nicotine upregulates microRNA-21 and promotes TGF-β-dependent epithelial-mesenchymal transition of esophageal cancer cells. Tumour Biol 2014; 35:7063-72. [PMID: 24756761 DOI: 10.1007/s13277-014-1968-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 04/11/2014] [Indexed: 12/15/2022] Open
Abstract
A consistent positive association between cigarette smoking and the human esophageal cancer has been confirmed all over the world. However, details in the association need to be more focused on and be identified. Recently, aberrantly expressed microRNAs (miRNAs) have been shown to be promising biomarkers for understanding the tumorigenesis of a wide array of human cancers, including the esophageal cancer, and the deregulation on the epithelial to mesenchymal transition (EMT) by miRNAs is involved in the tumorigenesis. In present study, we were going to identify the role of nicotine-induced miR-21 in the EMT of esophageal cells. We found that there was an overexpression of miR-21 in esophageal specimens, having an association with cigarette smoking, and the upregulation of miR-21 was also induced by nicotine in esophageal carcinoma cell line, EC9706. Moreover, the upregulated miR-21 by nicotine promoted EMT transforming growth factor beta (TGF-β) dependently. Thus, the present study reveals a novel oncogenic role of nicotine in human esophageal cancer.
Collapse
|