1
|
Liu S, Dai W, Jin B, Jiang F, Huang H, Hou W, Lan J, Jin Y, Peng W, Pan J. Effects of super-enhancers in cancer metastasis: mechanisms and therapeutic targets. Mol Cancer 2024; 23:122. [PMID: 38844984 PMCID: PMC11157854 DOI: 10.1186/s12943-024-02033-8] [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: 04/19/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Metastasis remains the principal cause of cancer-related lethality despite advancements in cancer treatment. Dysfunctional epigenetic alterations are crucial in the metastatic cascade. Among these, super-enhancers (SEs), emerging as new epigenetic regulators, consist of large clusters of regulatory elements that drive the high-level expression of genes essential for the oncogenic process, upon which cancer cells develop a profound dependency. These SE-driven oncogenes play an important role in regulating various facets of metastasis, including the promotion of tumor proliferation in primary and distal metastatic organs, facilitating cellular migration and invasion into the vasculature, triggering epithelial-mesenchymal transition, enhancing cancer stem cell-like properties, circumventing immune detection, and adapting to the heterogeneity of metastatic niches. This heavy reliance on SE-mediated transcription delineates a vulnerable target for therapeutic intervention in cancer cells. In this article, we review current insights into the characteristics, identification methodologies, formation, and activation mechanisms of SEs. We also elaborate the oncogenic roles and regulatory functions of SEs in the context of cancer metastasis. Ultimately, we discuss the potential of SEs as novel therapeutic targets and their implications in clinical oncology, offering insights into future directions for innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Shenglan Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 314000, China
| | - Wei Dai
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 314000, China
| | - Bei Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Feng Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 314000, China
| | - Hao Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 314000, China
| | - Wen Hou
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 314000, China
| | - Jinxia Lan
- College of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, China
| | - Yanli Jin
- College of Pharmacy, Jinan University Institute of Tumor Pharmacology, Jinan University, Guangzhou, 510632, China
| | - Weijie Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 314000, China.
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
2
|
Huang M, Hu J, Chen Y, Xun Y, Zhang X, Cao Y. Mesencephalic astrocyte-derived neurotrophic factor inhibits cervical cancer progression via regulating macrophage phenotype. Mol Biol Rep 2024; 51:654. [PMID: 38735002 DOI: 10.1007/s11033-024-09602-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Cervical cancer is a common gynecologic malignant tumor, but the critical factors affecting cervical cancer progression are still not well demonstrated. Mesencephalic astrocyte-derived neurotrophic factor (MANF) has been widely recognized as an anti-inflammatory factor to regulate macrophage polarization. In this study, the effect and mechanism of MANF on cervical cancer were preliminarily explored. METHODS AND RESULTS Kaplan-Meier curve was used to show the overall survival time of the involved cervical cancer patients with high and low MANF expression in cervical cancer tissues. MANF was highly expressed in peritumoral tissues of cervical carcinoma by using immunohistochemistry and western blot. MANF mRNA level was detected by using qRT-PCR. Dual-labeled immunofluorescence showed MANF was mainly expressed in macrophages of cervical peritumoral tissues. Moreover, MANF-silenced macrophages promoted HeLa and SiHa cells survival, migration, invasion and EMT via NF-κB signaling activation. The results of tumor formation in nude mice indicated MANF-silenced macrophages promoted cervical tumor formation in vivo. CONCLUSION Our study reveals an inhibitory role of MANF in cervical cancer progression, indicating MANF as a new and valuable therapeutic target for cervical cancer treatment.
Collapse
Affiliation(s)
- Miaomiao Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jingjing Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yueran Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Yingying Xun
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Xinru Zhang
- School of Basic Medical Sciences, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
3
|
Liang H, Zhao Y, Liu K, Xiao Y, Chen K, Li D, Zhong S, Zhao Z, Wu D, Peng Y. The mechanism of lncRNAs in the crosstalk between epithelial-mesenchymal transition and tumor microenvironment for early colon adenocarcinoma based on molecular subtyping. Front Genet 2022; 13:997739. [PMID: 36467998 PMCID: PMC9708740 DOI: 10.3389/fgene.2022.997739] [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: 07/19/2022] [Accepted: 10/17/2022] [Indexed: 09/10/2024] Open
Abstract
A large number of colon adenocarcinoma (COAD) patients are already advanced when diagnosed. In this study, we aimed to further understand the mechanism of tumor development in early COAD by focusing on epithelial-mesenchymal transition (EMT) and long non-coding RNAs (lncRNAs). Expression profiles of early COAD patients were obtained from public databases. EMT-related lncRNAs were used as a basis for constructing molecular subtypes through unsupervised consensus clustering. Genomic features, pathways and tumor microenvironment (TME) were compared between two subtypes. LncATLAS database was applied to analyze the relation between lncRNAs and transcription factors (TFs). First order partial correlation analysis was conducted to identify key EMT-related lncRNAs.C1 and C2 subtypes with distinct prognosis were constructed. Oncogenic pathways such as EMT, KRAS signaling, JAK-STAT signaling, and TGF-β signaling were significantly enriched in C2 subtype. Higher immune infiltration and expression of immune checkpoints were also observed in C2 subtype, suggesting the key EMT-related lncRNAs may play a critical role in the modulation of TME. In addition, JAK-STAT signaling pathway was obviously enriched in upregulated TFs in C2 subtype, which indicated a link between key lncRNAs and JAK-STAT signaling that may regulate TME. The study further expanded the research on the role of EMT-related lncRNAs in the early COAD. The six identified EMT-related lncRNAs could serve as biomarkers for early screening COAD.
Collapse
Affiliation(s)
- Hanlin Liang
- Chemotherapy Department, Zhongshan City People’s Hospital, Zhongshan, China
| | - Yi Zhao
- GI Medicine, The Third Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Kai Liu
- Department of Colorectal Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yajie Xiao
- Department of Medicine, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Kexu Chen
- Chemotherapy Department, Zhongshan City People’s Hospital, Zhongshan, China
| | - Delan Li
- Chemotherapy Department, Zhongshan City People’s Hospital, Zhongshan, China
| | - Shupeng Zhong
- Chemotherapy Department, Zhongshan City People’s Hospital, Zhongshan, China
| | - Zhikun Zhao
- Department of Medicine, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Dongfang Wu
- Department of Medicine, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Yu Peng
- Oncology Department, Jiangmen Central Hospital, Jiangmen, China
| |
Collapse
|
4
|
Piorecka K, Kurjata J, Stanczyk WA. Acriflavine, an Acridine Derivative for Biomedical Application: Current State of the Art. J Med Chem 2022; 65:11415-11432. [PMID: 36018000 PMCID: PMC9469206 DOI: 10.1021/acs.jmedchem.2c00573] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Acriflavine (ACF) has been known for years as an antibacterial
drug. The identification of key oncogenic mechanisms has brought,
in recent years, a significant increase in studies on ACF as a multipurpose
drug that would improve the prognosis for cancer patients. ACF interferes
with the expression of the hypoxia inducible factor, thus acting on
metastatic niches of tumors and significantly enhancing the effects
of other anticancer therapies. It has been recognized as the most
potent HIF-1 inhibitor out of the 336 drugs approved by the FDA. This
work presents up-to-date knowledge about the mechanisms of action
of ACF and its related prodrug systems in the context of anticancer
and SARS-CoV-2 inhibitory properties. It explains the multitask nature
of this drug and suggests mechanisms of ACF’s action on the
coronavirus. Other recent reports on ACF-based systems as potential
antibacterial and antiviral drugs are also described.
Collapse
Affiliation(s)
- Kinga Piorecka
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| | - Jan Kurjata
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| | - Wlodzimierz A Stanczyk
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| |
Collapse
|
5
|
Alba J, Barcia R, Gutiérrez-Berzal J, Ramos-Martínez JI. Could inhibition of metalloproteinases be used to block the process of metastasis? Cell Biochem Funct 2022; 40:600-607. [PMID: 35789101 PMCID: PMC9544369 DOI: 10.1002/cbf.3730] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 12/14/2022]
Abstract
Metastasis is a multisequential process that allows tumor cells to migrate to tissues distant from the primary tumor. Only a small number of cells escape from the primary tumor; however, the metastases generated are responsible for more than 90% of cancer deaths. Many metastatic processes initially require the total or partial start‐up of a program for the transformation of tumor epithelial cells into mesenchymal cells (EMT). The launching of the EMT program is stimulated by cytokines and other elements produced by the diverse types of cells composing the tumor stroma. In parallel, a process of destabilization of the extracellular matrix (ECM) takes place by means of the synthesis of proteases of the matrix metalloproteinases (MMPs) family. EMC degradation allows the exportation of some tumor cells as mesenchymal cells to the circulatory system and their subsequent implantation in a tissue distant from the primary tumor. The blocking of these both processes appears as a hypothetical stop point in the metastatic mechanism. The present review deals with the different options to achieve the inhibition of MMPs, focusing on MMP7 as a target given its involvement in the metastatic processes of a wide variety of tumors. The simultaneous implantation of the epithelial–mesenchymal program and the synthesis and activation of matrix metalloproteinases during the first phases of the metastasis process is known. The inhibition of proteases could constitute a possible blockage of the process. The review describes the evolution of the different inhibition mechanisms that could inform applicable therapeutic mechanisms for the paralysis of the metastatic process.
Collapse
Affiliation(s)
- Jesús Alba
- Histobiomol, Hospital POLUSA, Lugo, Spain
| | - Ramiro Barcia
- Faculty of Sciences, University of Santiago de Compostela, Lugo, Spain
| | | | - Juan I Ramos-Martínez
- Department of Biochemistry and Molecular Biology, School of Veterinary, University of Santiago de Compostela, Lugo, Spain
| |
Collapse
|
6
|
Zhang Y, Zhang K, Jia H, Xia B, Zang C, Liu Y, Qian L, Dong J. IVIM-DWI and MRI-based radiomics in cervical cancer: Prediction of concurrent chemoradiotherapy sensitivity in combination with clinical prognostic factors. Magn Reson Imaging 2022; 91:37-44. [PMID: 35568271 DOI: 10.1016/j.mri.2022.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE To identify the feasibility and value of intravoxel incoherent motion diffusion weighted imaging (IVIM-DWI) and magnetic resonance imaging (MRI)-based radiomics combined with clinical prognostic factors (CPF) in predicting concurrent chemoradiotherapy (CCRT) sensitivity of locally advanced cervical cancer (LACC). METHODS A retrospective analysis of 163 patients (assigned to training or test groups) who underwent conventional MRI and IVIM-DWI before CCRT were divided into sensitive and resistant groups according to their efficacy at 6 months after CCRT. Per-treatment IVIM-DWI parameters (ADC, D, D⁎ and f value), 3D texture features (from axial T2WI) and CPF were measured, analyzed and screened. The prediction model and its nomogram were developed by combining screened parameters and then validated internally and externally. RESULTS Clinical stage, f value, D value, InverseVariance, SizeZoneNonUniformity, and Minimum were selected to construct prediction model. All parameters except D value showed independent diagnostic value in multivariate Logistic regression analysis and composed prediction model, with AUCs of 0.987 and 0.984 for training and test groups, respectively. The calibration curve (Brier score of 0.042, C-index of 0.987), decision curve and clinical impact curve further demonstrated the reliability and clinical value of prediction model. CONCLUSION IVIM-DWI, MRI-based radiomics and CPF showed high clinical value in predicting CCRT sensitivity for LACC with better predictive performance when combined.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Radiation Oncology, Anhui Provincial Hospital Affiliated to Anhui Medical University, 17 Lujiang Road, Hefei, Anhui 230001, China
| | - Kaiyue Zhang
- Department of Radiation Oncology, Anhui Provincial Hospital Affiliated to Anhui Medical University, 17 Lujiang Road, Hefei, Anhui 230001, China
| | - Haodong Jia
- Department of Radiation Oncology, Anhui Provincial Hospital Affiliated to Anhui Medical University, 17 Lujiang Road, Hefei, Anhui 230001, China; Department of Radiology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China
| | - Bairong Xia
- Department of Radiology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China; Department of Radiation Oncology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China
| | - Chunbao Zang
- Department of Radiation Oncology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China
| | - Yunqin Liu
- Department of Radiation Oncology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China
| | - Liting Qian
- Department of Radiation Oncology, Anhui Provincial Hospital Affiliated to Anhui Medical University, 17 Lujiang Road, Hefei, Anhui 230001, China; Department of Radiation Oncology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China.
| | - Jiangning Dong
- Department of Radiology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China.
| |
Collapse
|
7
|
Wang H, Tang Y, Yang X, Wang W, Han P, Zhao J, He S, Liu P. A Crucial Angiogenesis-Associated Gene MEOX2 Could Be a Promising Biomarker Candidate for Breast Cancer. Front Oncol 2022; 12:759300. [PMID: 35615155 PMCID: PMC9124839 DOI: 10.3389/fonc.2022.759300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAngiogenesis plays a critical role in the growth and metastasis of breast cancer and angiogenesis inhibition has become an effective strategy for cancer therapy. Our study aimed to clarify the key candidate genes and pathways related to breast cancer angiogenesis.MethodsDifferentially expressed genes (DEGs) in the raw breast cancer (BRCA) gene dataset from the Cancer Genome Atlas (TCGA) database were identified and gene ontology analysis of the DEGs was performed. Hub genes were subsequently determined using the Gene Expression Omnibus database. The expression of the mesenchyme homeobox 2 (MEOX2) in breast cancer cells and tissues was assessed by quantification real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC), respectively. The prognostic value of the MEOX2 gene in breast cancer tissue was evaluated with the Kaplan-Meier plotter.ResultsA total of 61 angiogenesis-related DEGs were identified in the TCGA dataset, among which the gene MEOX2 was significantly down-regulated. GO functional annotation and pathway enrichment analyses showed that MEOX2 was significantly enriched in the regulation of vasculature development. The IHC results confirmed that MEOX2 expression was repressed in breast cancer tissues and the relatively low level indicated the tissue was densely vascularized. Moreover, MEOX2 expression was significantly elevated in breast cancer cells after treatment with cisplatin (DDP) and epirubicin (EPI). Finally, the Kaplan-Meier plotter confirmed that higher expression levels of MEOX2 were related to better overall survival.ConclusionOur study revealed that the angiogenesis-associated gene MEOX2 can be used as a novel biomarker for breast cancer diagnosis and clinical therapy.
Collapse
Affiliation(s)
- Huxia Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Yanan Tang
- Vascular Surgery Department, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaomin Yang
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Weiyi Wang
- Vascular Surgery Department, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Pihua Han
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Jing Zhao
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Sai He
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Peijun Liu,
| |
Collapse
|
8
|
Adiga D, Eswaran S, Pandey D, Sharan K, Kabekkodu SP. Molecular landscape of recurrent cervical cancer. Crit Rev Oncol Hematol 2020; 157:103178. [PMID: 33279812 DOI: 10.1016/j.critrevonc.2020.103178] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Cervical cancer (CC) is a major gynecological problem in developing and underdeveloped countries. Despite the significant advancement in early detection and treatment modalities, several patients recur. Moreover, the molecular mechanisms responsible for CC recurrence remains obscure. The patients with CC recurrence often show poor prognosis and significantly high mortality rates. The clinical management of recurrent CC depends on treatment history, site, and extent of the recurrence. Owing to poor prognosis and limited treatment options, recurrent CC often presents a challenge to the clinicians. Several in vitro, in vivo, and patient studies have led to the identification of the critical molecular changes responsible for CC recurrence. Both aberrant genetic and epigenetic modifications leading to altered cell signaling pathways have been reported to impact CC recurrence. Researchers are currently trying to dissect the molecular pathways in CC and translate these findings for better management of disease. This article attempts to review the existing knowledge of disease relapse, accompanying challenges, and associated molecular players in CC.
Collapse
Affiliation(s)
- Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sangavi Eswaran
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Deeksha Pandey
- Department of OBGYN, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishna Sharan
- Department of Radiotherapy and Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
9
|
Xiong W, Liao Y, Qin JY, Li WH, Tang ZY. Adverse effects of chemoradiotherapy on invasion and metastasis of tumor cells. Genes Dis 2020; 7:351-358. [PMID: 32884989 PMCID: PMC7452502 DOI: 10.1016/j.gendis.2020.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/18/2020] [Accepted: 04/03/2020] [Indexed: 12/21/2022] Open
Abstract
The phenomenon of enhanced invasion and metastasis of residual tumor cells has been observed in an increasing number of patients receiving chemoradiotherapy recently, and tumor metastasis will undoubtedly limit patient prognosis. However, the key mechanism by which chemoradiotherapy affects the invasion and metastasis of tumor cells remains unclear. Studies have shown that chemoradiotherapy may directly act on tumor cells and alter the tumor microenvironment, or induce cell apoptosis and autophagy to promote tumor cell survival and metastasis. In this review, we summarize the potential mechanisms by which chemoradiotherapy may affect the biological behavior of tumor cells and open up new avenues for reducing tumor recurrence and metastasis after treatment. These insights will improve the efficacy of chemoradiotherapy.
Collapse
Affiliation(s)
- Wei Xiong
- The Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yong Liao
- Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ji-Yong Qin
- The Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wen-Hui Li
- The Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao-You Tang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Estradiol Induces Epithelial to Mesenchymal Transition of Human Glioblastoma Cells. Cells 2020; 9:cells9091930. [PMID: 32825553 PMCID: PMC7564468 DOI: 10.3390/cells9091930] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/02/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
The mesenchymal phenotype of glioblastoma multiforme (GBM), the most frequent and malignant brain tumor, is associated with the worst prognosis. The epithelial–mesenchymal transition (EMT) is a cell plasticity mechanism involved in GBM malignancy. In this study, we determined 17β-estradiol (E2)-induced EMT by changes in cell morphology, expression of EMT markers, and cell migration and invasion assays in human GBM-derived cell lines. E2 (10 nM) modified the shape and size of GBM cells due to a reorganization of actin filaments. We evaluated EMT markers expression by RT-qPCR, Western blot, and immunofluorescence.We found that E2 upregulated the expression of the mesenchymal markers, vimentin, and N-cadherin. Scratch and transwell assays showed that E2 increased migration and invasion of GBM cells. The estrogen receptor-α (ER-α)-selective agonist 4,4’,4’’-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT, 10 nM) affected similarly to E2 in terms of the expression of EMT markers and cell migration, and the treatment with the ER-α antagonist methyl-piperidino-pyrazole (MPP, 1 μM) blocked E2 and PPT effects. ER-β-selective agonist diarylpropionitrile (DNP, 10 nM) and antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazole[1,5-a]pyrimidin-3-yl]phenol (PHTPP, 1 μM) showed no effects on EMT marker expression. These data suggest that E2 induces EMT activation through ER-α in human GBM-derived cells.
Collapse
|
11
|
Yang SS, Yu DY, Du YT, Wang L, Gu L, Zhang YY, Xiao M. Inhibition of Delta-like Ligand 4 enhances the radiosensitivity and inhibits migration in cervical cancer via the reversion of epithelial-mesenchymal transition. Cancer Cell Int 2020; 20:344. [PMID: 32742191 PMCID: PMC7388465 DOI: 10.1186/s12935-020-01434-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 07/18/2020] [Indexed: 12/24/2022] Open
Abstract
Background Concurrent chemoradiotherapy is the common first-line treatment for patients with advanced cervical cancer. However, radioresistance remains a major clinical challenge, which results in recurrence and poor survival. Many studies have shown the potential of Delta-like Ligand 4 (DLL4) as a novel prognostic biomarker and therapeutic target in many solid tumors. Previously, we have found that high DLL4 expression in tumor cells may predict the pelvic lymph node metastasis and poor prognosis in patients with cervical cancer. In our present study, we further studied the effects of DLL4 on the biological behavior and radiosensitivity of cervical cancer cells. Methods The expression of DLL4 and epithelial–mesenchymal transition (EMT) phenotype markers in cervical cancer cell lines or tissues were detected using Western blotting, and the expression of DLL4 mRNA in cervical cancer cell lines or tissues was detected using Quantitative real-time PCR. The effect of DLL4 on cell proliferation, migration, and radiosensitivity was evaluated using the CCK8 assay, flow cytometry, Transwell assays for cell invasion and migration, and Immunofluorescence staining in vitro. Results The expression of DLL4 in radiotherapy-resistant SiHa cells was significantly higher than that in radiotherapy-sensitive Me-180 cells. Furthermore, downregulation of DLL4 enhanced the radiosensitivity of SiHa and Caski cells via the inhibition of cell proliferation, promotion of radiation-induced apoptosis, and inhibition of the DNA damage repair. Moreover, downregulation of DLL4 inhibited the EMT and reduced the proliferation, invasion, and migration ability in SiHa and Caski cells. Consistent with the DLL4 expression in the cell lines, the expression of DLL4 in the tissues of the radioresistant group was also higher than that of the radiosensitive group. Conclusions Downregulation of DLL4 inhibited the progression and increased the radiosensitivity in cervical cancer cells by reversing EMT. These results indicated the promising prospect of DLL4 against the radioresistance and metastasis of cervical cancer and its potential as a predictive biomarker for radiosensitivity and prognosis in patients with cervical cancer patients receiving concurrent chemoradiotherapy (cCRT).
Collapse
Affiliation(s)
- Shan-Shan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, No. 150 HaPing Road, Nangang District, Harbin, 150081 China
| | - De-Yang Yu
- Department of Radiation Physics, Harbin Medical University Cancer Hospital, Harbin, 150081 China
| | - Yu-Ting Du
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, No. 150 HaPing Road, Nangang District, Harbin, 150081 China
| | - Le Wang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, No. 150 HaPing Road, Nangang District, Harbin, 150081 China
| | - Lina Gu
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, No. 150 HaPing Road, Nangang District, Harbin, 150081 China
| | - Yun-Yan Zhang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, No. 150 HaPing Road, Nangang District, Harbin, 150081 China
| | - Min Xiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150 HaPing Road, Nangang District, Harbin, 150081 China
| |
Collapse
|
12
|
Ribatti D, Tamma R, Annese T. Epithelial-Mesenchymal Transition in Cancer: A Historical Overview. Transl Oncol 2020; 13:100773. [PMID: 32334405 PMCID: PMC7182759 DOI: 10.1016/j.tranon.2020.100773] [Citation(s) in RCA: 478] [Impact Index Per Article: 119.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/26/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transitions (EMTs), the acquisition of mesenchymal features from epithelial cells, occur during some biological processes and are classified into three types: the first type occurs during embryonic development, the second type is associated with adult tissue regeneration, and the third type occurs in cancer progression. EMT occurring during embryonic development in gastrulation, renal development, and the origin and fate of the neural crest is a highly regulated process, while EMT occurring during tumor progression is highly deregulated. EMT allows the solid tumors to become more malignant, increasing their invasiveness and metastatic activity. Secondary tumors frequently maintain the typical histologic characteristics of the primary tumor. These histologic features connecting the secondary metastatic tumors to the primary is due to a process called mesenchymal-epithelial transition (MET). MET has been demonstrated in different mesenchymal tumors and is the expression of the reversibility of EMT. EMT modulation could constitute an approach to avoid metastasis. Some of the targeted small molecules utilized as antiproliferative agents have revealed to inhibit EMT initiation or maintenance because EMT is regulated through signaling pathways for which these molecules have been designed.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| |
Collapse
|
13
|
Suhail Y, Cain MP, Vanaja K, Kurywchak PA, Levchenko A, Kalluri R, Kshitiz. Systems Biology of Cancer Metastasis. Cell Syst 2019; 9:109-127. [PMID: 31465728 PMCID: PMC6716621 DOI: 10.1016/j.cels.2019.07.003] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/29/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022]
Abstract
Cancer metastasis is no longer viewed as a linear cascade of events but rather as a series of concurrent, partially overlapping processes, as successfully metastasizing cells assume new phenotypes while jettisoning older behaviors. The lack of a systemic understanding of this complex phenomenon has limited progress in developing treatments for metastatic disease. Because metastasis has traditionally been investigated in distinct physiological compartments, the integration of these complex and interlinked aspects remains a challenge for both systems-level experimental and computational modeling of metastasis. Here, we present some of the current perspectives on the complexity of cancer metastasis, the multiscale nature of its progression, and a systems-level view of the processes underlying the invasive spread of cancer cells. We also highlight the gaps in our current understanding of cancer metastasis as well as insights emerging from interdisciplinary systems biology approaches to understand this complex phenomenon.
Collapse
Affiliation(s)
- Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, USA; Cancer Systems Biology @ Yale (CaSB@Yale), Yale University, West Haven, CT, USA
| | - Margo P Cain
- Department of Cancer Biology, MD Anderson Cancer Center, Houston, TX, USA
| | - Kiran Vanaja
- Cancer Systems Biology @ Yale (CaSB@Yale), Yale University, West Haven, CT, USA
| | - Paul A Kurywchak
- Department of Cancer Biology, MD Anderson Cancer Center, Houston, TX, USA
| | - Andre Levchenko
- Cancer Systems Biology @ Yale (CaSB@Yale), Yale University, West Haven, CT, USA
| | - Raghu Kalluri
- Department of Cancer Biology, MD Anderson Cancer Center, Houston, TX, USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, USA; Cancer Systems Biology @ Yale (CaSB@Yale), Yale University, West Haven, CT, USA.
| |
Collapse
|
14
|
Das K, Paul S, Singh A, Ghosh A, Roy A, Ansari SA, Prasad R, Mukherjee A, Sen P. Triple-negative breast cancer-derived microvesicles transfer microRNA221 to the recipient cells and thereby promote epithelial-to-mesenchymal transition. J Biol Chem 2019; 294:13681-13696. [PMID: 31341019 DOI: 10.1074/jbc.ra119.008619] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/08/2019] [Indexed: 12/15/2022] Open
Abstract
The triple-negative phenotype is the most prevalent form of human breast cancer worldwide and is characterized by poor survival, high aggressiveness, and recurrence. Microvesicles (MV) are shredded plasma membrane components and critically mediate cell-cell communication, but can also induce cancer proliferation and metastasis. Previous studies have revealed that protease-activated receptor 2 (PAR2) contributes significantly to human triple-negative breast cancer (TNBC) progression by releasing nano-size MV and promoting cell proliferation, migration, and invasion. MV isolated from highly aggressive human TNBC cells impart metastatic potential to nonmetastatic cells. Over-expression of microRNA221 (miR221) has also been reported to enhance the metastatic potential of human TNBC, but miR221's relationship to PAR2-induced MV is unclear. Here, using isolated MV, immunoblotting, quantitative RT-PCR, FACS analysis, and enzymatic assays, we show that miR221 is translocated via human TNBC-derived MV, which upon fusion with recipient cells, enhance their proliferation, survival, and metastasis both in vitro and in vivo by inducing the epithelial-to-mesenchymal transition (EMT). Administration of anti-miR221 significantly impaired MV-induced expression of the mesenchymal markers Snail, Slug, N-cadherin, and vimentin in the recipient cells, whereas restoring expression of the epithelial marker E-cadherin. We also demonstrate that MV-associated miR221 targets phosphatase and tensin homolog (PTEN) in the recipient cells, followed by AKT Ser/Thr kinase (AKT)/NF-κB activation, which promotes EMT. Moreover, elevated miR221 levels in MV derived from human TNBC patients' blood could induce cell proliferation and metastasis in recipient cells. In summary, miR221 transfer from TNBC cells via PAR2-derived MV induces EMT and enhances the malignant potential of recipient cells.
Collapse
Affiliation(s)
- Kaushik Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Arpana Singh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Arnab Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Abhishek Roy
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | | | - Ramesh Prasad
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Ashis Mukherjee
- A Unit of Himadri Memorial Cancer Welfare Trust, Netaji Subhash Chandra Bose Cancer Research Institute, Kolkata 700016, India
| | - Prosenjit Sen
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
15
|
Transglutaminase 2 maintains a colorectal cancer stem phenotype by regulating epithelial-mesenchymal transition. Oncotarget 2019; 10:4556-4569. [PMID: 31360304 PMCID: PMC6642042 DOI: 10.18632/oncotarget.27062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
Transglutaminase 2 (TG2), a multifunctional protein, is reported in regulating the cancer stem cell (CSC) phenotype in various cancers. Our previous work suggested the link between TG2 and Epithelial-Mesenchymal Transition (EMT) in colorectal cancer (CRC). Here we demonstrate the importance of TG2 in CSC development in human CRC cell lines HCT116 and SW620. CRC spheroid cells showed increased CSC characteristics over their monolayer cells with increased expression of CD44 and over expression of Oct3/4, Sox2 and Nanog. They also showed increased EMT and invasiveness, and enhanced expression of TG2. TG2 inhibition by its selective inhibitor 1-155 reduced both spheroid formation and invasive potential of the spheroid cells. β-catenin, a mediator of stem cell maintenance, was overexpressed in the spheroid cells and could be attenuated by TG2 inhibition. Spheroid cells possessed increased angiogenesis stimulating ability via overexpression of Vascular Endothelial Growth Factor (VEGF). Increased VEGF was present in the culture media from spheroid cells when compared to monolayer cultures which could be reduced by selective inhibition by 1-155. Stemness and malignancy in the colorectal spheroid cells was associated with increased TG2, EMT, β-catenin and VEGF. Here we demonstrate that inhibiting TG2 reduces both stemness and angiogenic stimulating activity in CRC.
Collapse
|
16
|
Jaskiewicz NM, Townson DH. Hyper-O-GlcNAcylation promotes epithelial-mesenchymal transition in endometrial cancer cells. Oncotarget 2019; 10:2899-2910. [PMID: 31080560 PMCID: PMC6499600 DOI: 10.18632/oncotarget.26884] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/03/2019] [Indexed: 01/06/2023] Open
Abstract
Diabetic women have a 2–3 fold increased risk of developing endometrial cancer, however, the molecular aspects of this risk are not fully understood. This study investigated the alteration of cellular O-GlcNAcylation of proteins as the potential mechanistic connection between these two conditions. The endometrial cancer cell line (Ishikawa) was utilized to study the effect of dysregulation of O-GlcNAcylation on epithelial mesenchymal transition (EMT). Hyper-O-GlcNAcylation (via 1 μM Thiamet-G/ThmG or 25 mM Glucose) enhanced the expression of EMT-associated genes (WNT5B and FOXC2), and protein expression of the EMT adhesion molecule, N-Cadherin. Reorganization of stress filaments (actin filaments), consistent with EMT, was also noted in ThmG-treated cells. Interestingly, Hypo-O-GlcNAcylation (via 50 μM OSMI-1) also upregulated WNT5B, inferring that any disruption to O-GlcNAc cycling impacts EMT. However, Hypo-O-GlcNAcylation reduced overall cellular proliferation/migration and the expression of pro-EMT genes (AHNAK, TGFB2, FGFBP1, CALD1, TFPI2). In summary, disruption of O-GlcNAc cycling (i.e., Hyper- or Hypo-O-GlcNAcylation) promoted EMT at both the molecular and cellular levels, but only Hyper-O-GlcNAcylation provoked cellular proliferation/migration, and cytoskeletal reorganization.
Collapse
Affiliation(s)
- Nicole Morin Jaskiewicz
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - David H Townson
- Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT, USA
| |
Collapse
|
17
|
Pang Y, Mao SS, Yao R, He JY, Zhou ZZ, Feng L, Zhang KT, Cheng SJ, Sun W. TGF-β induced epithelial-mesenchymal transition in an advanced cervical tumor model by 3D printing. Biofabrication 2018; 10:044102. [PMID: 30129928 DOI: 10.1088/1758-5090/aadbde] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
An advanced in vitro cervical tumor model was established by 3D printing to study the epithelial-to-mesenchymal transition (EMT), which is a very important stage of dissemination of carcinoma leading to metastatic tumors. A HeLa/hydrogel grid construct composed of gelatin, alginate, Matrigel and HeLa cells was fabricated by forced extrusion in a layer-by-layer fashion. HeLa cells rapidly proliferated, formed spheroids and presented tumorigenic characteristic in the 3D-printed structure. With the supplement of TGF-β, aggregated HeLa cells started to disintegrate, and some of them changed into fibroblast-like spindle morphology, which indicated that EMT was induced. The down-regulation of epithelial marker E-cadherin, and up-regulation of mesenchymal markers such as snail, vimentin and N-cadherin were all observed in the 3D-printed model, and performed differently in 3D and 2D models. The TGF-β induced EMT was inhibited by the treatment of disulfiram and EMT pathway inhibitor C19 in a dose dependent manner, showing great potential for future studies of a therapeutic program towards cervical tumor metastasis.
Collapse
Affiliation(s)
- Y Pang
- Biomanufacturing Center, Dept. of Mechanical Engineering, Tsinghua University, Haidian District, Beijing 100084, People's Republic of China. Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China. Overseas Expertise Introduction Center for Discipline Innovation, Tsinghua University, Haidian District, Beijing 100084, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Li J, Khan MA, Wei C, Cheng J, Chen H, Yang L, Ijaz I, Fu J. Thymoquinone Inhibits the Migration and Invasive Characteristics of Cervical Cancer Cells SiHa and CaSki In Vitro by Targeting Epithelial to Mesenchymal Transition Associated Transcription Factors Twist1 and Zeb1. Molecules 2017; 22:molecules22122105. [PMID: 29207526 PMCID: PMC6149891 DOI: 10.3390/molecules22122105] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 11/16/2022] Open
Abstract
Cervical cancer is one of the most common gynecological malignant tumors worldwide, for which chemotherapeutic strategies are limited due to their non-specific cytotoxicity and drug resistance. The natural product thymoquinone (TQ) has been reported to target a vast number of signaling pathways in carcinogenesis in different cancers, and hence is regarded as a promising anticancer molecule. Inhibition of epithelial to mesenchymal transition (EMT) regulators is an important approach in anticancer research. In this study, TQ was used to treat the cervical cancer cell lines SiHa and CaSki to investigate its effects on EMT-regulatory proteins and cancer metastasis. Our results showed that TQ has time-dependent and dose-dependent cytotoxic effects, and it also inhibits the migration and invasion processes in different cervical cancer cells. At the molecular level, TQ treatment inhibited the expression of Twist1, Zeb1 expression, and increased E-Cadherin expression. Luciferase reporter assay showed that TQ decreases the Twist1 and Zeb1 promoter activities respectively, indicating that Twist1 and Zeb1 might be the direct target of TQ. TQ also increased cellular apoptosis in some extent, but apoptotic genes/proteins we tested were not significant affected. We conclude that TQ inhibits the migration and invasion of cervical cancer cells, probably via Twist1/E-Cadherin/EMT or/and Zeb1/E-Cadherin/EMT, among other signaling pathways.
Collapse
Affiliation(s)
- Jun Li
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China.
| | - Md Asaduzzaman Khan
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China.
| | - Chunli Wei
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China.
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China.
| | - Hanchun Chen
- Department of Biochemistry, School of Life Sciences, Central South University, Changsha 410013, China.
| | - Lisha Yang
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China.
| | - Iqra Ijaz
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China.
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China.
- Medical College, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
19
|
Li W, Cai S, Wang L, Yang C, Zhou B, Wang H. HINT2 downregulation promotes colorectal carcinoma migration and metastasis. Oncotarget 2017; 8:13521-13531. [PMID: 28088787 PMCID: PMC5355117 DOI: 10.18632/oncotarget.14587] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 01/03/2017] [Indexed: 01/05/2023] Open
Abstract
Histidine triad nucleotide-binding 2 (HINT2), a member of the histidine triad proteins family, sensitizes cells to apoptosis in hepatocellular carcinoma. Here, we showed that HINT2 expression is lower in primary colorectal cancer (CRC) and metastasis tissues than in normal colorectal tissues, and that HINT2 abundance is inversely correlated with CRC tumor stage. Treating CRC cells with 5-aza-2′-deoxycytidine, a demethylating agent, upregulated HINT2, suggesting HINT2 downregulation is caused by methylation of the gene promoter. HINT2 downregulation increased tumor migration and invasion in vitro, promoted CRC cell metastasis in vivo, and increased expression of epithelial-to-mesenchymal transition (EMT) markers. Furthermore, HINT2 downregulation depended on hypoxia inducible factor (HIF)-2α-mediated transcriptional activation of zinc finger E-box-binding homeobox 1 (ZEB1). These results suggest that HINT2 downregulation promotes HIF-2α expression, which induces EMT and enhances CRC cell migration and invasion. HINT2 may thus a useful clinical indicator of CRC progression and metastasis risk.
Collapse
Affiliation(s)
- Weihua Li
- Department of Surgical Oncology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou 350001, China
| | - Shaoxin Cai
- Department of Surgical Oncology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou 350001, China
| | - Le Wang
- Department of Surgical Oncology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou 350001, China
| | - Changshun Yang
- Department of Surgical Oncology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou 350001, China
| | - Biaohuan Zhou
- Department of Surgical Oncology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou 350001, China
| | - Huan Wang
- Department of Surgical Oncology, Fujian Provincial Clinical College, Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
20
|
Prognostic value of the pretreatment neutrophil-to-lymphocyte ratio in cervical cancer: a meta-analysis and systematic review. Oncotarget 2017; 8:13400-13412. [PMID: 28077792 PMCID: PMC5355107 DOI: 10.18632/oncotarget.14541] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/28/2016] [Indexed: 12/21/2022] Open
Abstract
The prognostic value of pretreatment neutrophil-to-lymphocyte ratio (NLR) in cervical cancer remains controversial. We conducted a meta-analysis based on the data from 13 studies with 3729 patients to evaluate the association between the pretreatment NLR and the clinical outcomes of overall survival and progression-free survival in patients with cervical cancer. The relationship between NLR and clinicopathological parameters was also assessed. Hazard ratio (HR) or odds ratio (OR) with its 95% confidence interval (CI) was used as the effect size estimate. Our analysis indicated that elevated pretreatment NLR was a poor prognostic marker for patients with cervical cancer because it predicted unfavorable overall survival (HR = 1.375, 95% CI: 1.200–1.576) and progression-free survival (HR = 1.646, 95% CI: 1.313–2.065). Increased NLR is also significantly associated with the larger tumor size (OR = 1.780, 95% CI: 1.090–2.908), advanced clinical stage (OR = 2.443, 95% CI: 1.730–3.451), and positive lymph node metastasis (OR = 2.380, 95% CI: 1.775–3.190). By these results, high pretreatment NLR predicted a shorter survival period for patients with cervical cancer, and it could be served as a novel index of prognostic evaluation in patients with cervical cancer.
Collapse
|
21
|
Short interfering RNA targeting Net1 reduces the angiogenesis and tumor growth of in vivo cervical squamous cell carcinoma through VEGF down-regulation. Hum Pathol 2017; 65:113-122. [DOI: 10.1016/j.humpath.2017.04.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/18/2017] [Accepted: 04/29/2017] [Indexed: 12/22/2022]
|
22
|
Sheng SR, Wu JS, Tang YL, Liang XH. Long noncoding RNAs: emerging regulators of tumor angiogenesis. Future Oncol 2017; 13:1551-1562. [PMID: 28513194 DOI: 10.2217/fon-2017-0149] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) participate in multiple biological processes especially human diseases, of which, tumor seems to be one of the most significant. Angiogenesis has been deemed to have a pivotal role in a series of tumor biological behaviors in tumorigenesis, progression and prognosis. Emerging evidences suggested that lncRNAs are involved in tumor angiogenesis and lncRNAs have already been verified to be potential biomarkers and promising therapeutic targets. This review summarized emerging angiogenesis-related lncRNAs, discussed their mechanisms interacting with cytokines, cancer stem cells, miRNAs and tumor hypoxia microenvironment, and demonstrated if lncRNAs could be new candidate targets of antiangiogenesis therapy.
Collapse
Affiliation(s)
- Su-Rui Sheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China.,Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China
| | - Jia-Shun Wu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China.,Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China.,Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China.,Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
23
|
Garg M. Epithelial, mesenchymal and hybrid epithelial/mesenchymal phenotypes and their clinical relevance in cancer metastasis. Expert Rev Mol Med 2017; 19:e3. [PMID: 28322181 DOI: 10.1017/erm.2017.6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer metastasis occurs through local invasion of circulating tumour cells (CTCs), intravasation, transportation to distant sites, and their extravasation followed by colonisation at secondary sites. Epithelial-mesenchymal transition (EMT) is a normal developmental phenomenon, but its aberrant activation confers tumour cells with enhanced cell motility, metastatic properties, resistant to therapies and cancer stem cell (CSC) phenotype in epithelium-derived carcinoma. Experimental studies from various research papers have been reviewed to determine the factors, which interlink cancer stemness and cellular plasticity with EMT. Although existence of CSCs has been linked with EMT, nevertheless, there are controversies with the involvement of type of tumour cells, including cells with E (epithelial) and M (mesenchymal) phenotype alone or hybrid E/M phenotype in different types of cancers. Studies on CTCs with hybrid E/M phenotypes during different stages of cancer metastasis reveal strong association with tumour -initiation potential, cellular plasticity and types of cancer cells. Cells with the hybrid E/M state are strictly controlled by phenotypic stability factors coupled to core EMT decision-making circuits, miR200/ZEB and miR-34/Snail. Understanding the regulatory functions of EMT program in cancer metastasis can help us to characterise the biomarkers of prognostic and therapeutic potential. These biomarkers when targeted may act as metastatic suppressors, inhibit cellular plasticity and stemness ability of tumour cells and can block metastatic growth.
Collapse
Affiliation(s)
- Minal Garg
- Department of Biochemistry,University of Lucknow,Lucknow - 226007,UP,India
| |
Collapse
|
24
|
ADC Histogram Analysis of Cervical Cancer Aids Detecting Lymphatic Metastases—a Preliminary Study. Mol Imaging Biol 2017; 19:953-962. [DOI: 10.1007/s11307-017-1073-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
25
|
Epithelial-mesenchymal transition in morphogenesis, cancer progression and angiogenesis. Exp Cell Res 2017; 353:1-5. [PMID: 28257786 DOI: 10.1016/j.yexcr.2017.02.041] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 12/18/2022]
Abstract
All organs consist of an epithelium and an associated mesenchyme, so these epithelial-mesenchymal intercations are among the most important phenomena in nature. The aim of this article is the summarize the common mechanisms involved in the establishment of epithelial mesenchymal transition in three biological processes, namely organogenesis, tumor progression and metastasis, and angiogenesis, apparently independent each from other. A common feature of these processes is the fact that specialized epithelial cells lose their features, including cell adhesion and polarity, reorganize their cytoskeleton, and acquire a mesenchymal morphology and the ability to migrate.
Collapse
|
26
|
Rojas-Puentes L, Cardona AF, Carranza H, Vargas C, Jaramillo LF, Zea D, Cetina L, Wills B, Ruiz-Garcia E, Arrieta O. Epithelial-mesenchymal transition, proliferation, and angiogenesis in locally advanced cervical cancer treated with chemoradiotherapy. Cancer Med 2016; 5:1989-99. [PMID: 27230280 PMCID: PMC4884920 DOI: 10.1002/cam4.751] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/09/2016] [Accepted: 03/25/2016] [Indexed: 12/23/2022] Open
Abstract
We evaluated the association between epithelial-mesenchymal transition (EMT)-derived markers and expression of proteins associated with cell proliferation and tumor growth, as well as their prognostic roles, in 61 patients (mean age 52 ± 10 years) with locally advanced cervical cancer, all of whom were treated with chemoradiation and intracavitary brachytherapy. We used immunohistochemical analysis to assess the expression of proteins targeted in our investigation. Various statistical analyses were then conducted to assess protein marker associations with survival outcomes. Forty-six percent of the patients were positive for human papilloma virus. Median progression-free survival (PFS) was 6.6 months (95% confidence interval [CI]: 4.0-9.1, whereas overall survival (OS) was 30.0 months (95% CI: 11-48). Multivariate analysis demonstrated that vascular endothelial growth factor (VEGF) (P = 0.002), epidermal growth factor receptor (EGFR) (P = 0.001), and TWIST2 (P = 0.001) expression levels, as well as a tumor size <6 cm (P = 0.02), influenced OS. Changes in TWIST2 levels and loss of E-cadherin expression were correlated with VEGF and EGFR levels; furthermore, patients with high TWIST2 expression had shorter OS (P = 0.0001), as those with loss of E-cadherin (P = 0.02). OS was even shorter when positive EGFR or VEGF expression was related with EMT markers (positive EGFR + negative E-cadherin: median 14 months, 95% CI: 3-24; negative EGFR + positive E-cadherin: median 31 months, 95% CI: 14-NA; P = 0.02.). The presence of EMT markers was associated with proliferative and pro-angiogenic protein expression and influenced the prognosis of locally advanced cervical cancer.
Collapse
Affiliation(s)
- Leonardo Rojas-Puentes
- Clinical Oncology Group, Centro Javeriano de Oncología, Hospital Universitario San Ignacio (HUSI), Bogotá, Colombia
| | - Andrés F Cardona
- Clinical and Translational Oncology Group, Institute of Oncology, Clínica del Country, Bogotá, Colombia
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia
| | - Hernán Carranza
- Clinical and Translational Oncology Group, Institute of Oncology, Clínica del Country, Bogotá, Colombia
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia
| | - Carlos Vargas
- Clinical and Translational Oncology Group, Institute of Oncology, Clínica del Country, Bogotá, Colombia
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia
| | - Luis F Jaramillo
- Department of Pathology, Hospital Universitario San Ignacio (HUSI), Bogotá, Colombia
| | - Delma Zea
- Radiotherapy Group, Centro Javeriano de Oncología, Hospital Universitario San Ignacio (HUSI), Bogotá, Colombia
| | - Lucely Cetina
- Department of Medical Oncology, Instituto Nacional de Cancerología de México (INCAN), México City, México
| | - Beatriz Wills
- Clinical and Translational Oncology Group, Institute of Oncology, Clínica del Country, Bogotá, Colombia
| | - Erika Ruiz-Garcia
- Department of Medical Oncology, Instituto Nacional de Cancerología de México (INCAN), México City, México
| | - Oscar Arrieta
- Department of Medical Oncology, Instituto Nacional de Cancerología de México (INCAN), México City, México
| |
Collapse
|