1
|
Xue H, Han L, Sun H, Piao Z, Cao W, Qian H, Zhao Z, Lang MF, Gu C. Metastasis-associated 1 localizes to the sarcomeric Z-disc and is implicated in skeletal muscle pathology. Cytoskeleton (Hoboken) 2024; 81:427-435. [PMID: 38391059 DOI: 10.1002/cm.21841] [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: 07/19/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024]
Abstract
Metastasis-associated 1 (MTA1), a subunit of the nucleosome remodeling and histone deacetylation (NuRD) corepressor complex, was reported to be expressed in the cytoplasm of skeletal muscles. However, the exact subcellular localization and the functional implications of MTA1 in skeletal muscles have not been examined. This study aims to demonstrate the subcellular localization of MTA1 in skeletal muscles and reveal its possible roles in skeletal muscle pathogenesis. Striated muscles (skeletal and cardiac) from C57BL/6 mice of 4-5 weeks were collected to examine the expression of MTA1 by Western blotting and immunohistochemistry. Immunofluorescence and immunoelectron microscopy were performed for MTA1, α-actinin (a Z-disc marker protein), and SMN (survival of motor neuron) proteins. Gene Expression Omnibus (GEO) data sets were analyzed using the GEO2R online tool to explore the functional implications of MTA1 in skeletal muscles. MTA1 expression was detected by Western blotting and immunohistochemistry in skeletal and cardiac muscles. Subcellular localization of MTA1 was found in the Z-disc of sarcomeres, where α-actinin and SMN were expressed. Data mining of GEO profiles suggested that MTA1 dysregulation is associated with multiple skeletal muscle defects, such as Duchenne muscular dystrophy, Emery-Dreifuss muscular dystrophy, nemaline myopathy, and dermatomyositis. The GEO analysis also showed that MTA1 expression gradually decreased with age in mouse skeletal muscle precursor cells. The subcellular localization of MTA1 in sarcomeres of skeletal muscles implies its biological roles in sarcomere structures and its possible contribution to skeletal muscle pathology.
Collapse
Affiliation(s)
- Hongsheng Xue
- Department of Thoracic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Li Han
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Haidi Sun
- Department of Physiology and Medical Biology, Medical College, Dalian University, Dalian, China
| | - Zhe Piao
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Wenjun Cao
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhilong Zhao
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ming-Fei Lang
- Department of Physiology and Medical Biology, Medical College, Dalian University, Dalian, China
| | - Chundong Gu
- Department of Thoracic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
2
|
Iyer AS, Shaik MR, Raufman JP, Xie G. The Roles of Zinc Finger Proteins in Colorectal Cancer. Int J Mol Sci 2023; 24:10249. [PMID: 37373394 DOI: 10.3390/ijms241210249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Despite colorectal cancer remaining a leading worldwide cause of cancer-related death, there remains a paucity of effective treatments for advanced disease. The molecular mechanisms underlying the development of colorectal cancer include altered cell signaling and cell cycle regulation that may result from epigenetic modifications of gene expression and function. Acting as important transcriptional regulators of normal biological processes, zinc finger proteins also play key roles in regulating the cellular mechanisms underlying colorectal neoplasia. These actions impact cell differentiation and proliferation, epithelial-mesenchymal transition, apoptosis, homeostasis, senescence, and maintenance of stemness. With the goal of highlighting promising points of therapeutic intervention, we review the oncogenic and tumor suppressor roles of zinc finger proteins with respect to colorectal cancer tumorigenesis and progression.
Collapse
Affiliation(s)
- Aishwarya S Iyer
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mohammed Rifat Shaik
- Department of Medicine, University of Maryland Medical Center Midtown Campus, Baltimore, MD 21201, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Guofeng Xie
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
Feng M, Liu L, Qu Z, Zhang B, Wang Y, Yan L, Kong L. CRISPR/Cas9 knockout of MTA1 enhanced RANKL-induced osteoclastogenesis in RAW264.7 cells partly via increasing ROS activities. J Cell Mol Med 2023; 27:701-713. [PMID: 36786127 PMCID: PMC9983315 DOI: 10.1111/jcmm.17692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/16/2023] [Accepted: 01/29/2023] [Indexed: 02/15/2023] Open
Abstract
Metastasis-associated protein 1 (MTA1), belonging to metastasis-associated proteins (MTA) family, which are integral parts of nucleosome remodelling and histone deacetylation (NuRD) complexes. However, the effect of MTA1 on osteoclastogenesis is unknown. Currently, the regulation of MTA1 in osteoclastogenesis was reported for the first time. MTA1 knockout cells (KO) were established by CRISPR/Cas9 genome editing. RAW264.7 cells with WT and KO group were stimulated independently by RANKL to differentiate into mature osteoclasts. Further, western blotting and quantitative qRT-PCR were used to explore the effect of MTA1 on the expression of osteoclast-associated genes (including CTSK, MMP9, c-Fos and NFATc1) during osteoclastogenesis. Moreover, the effects of MTA1 on the expression of reactive oxygen species (ROS) in osteoclastogenesis was determined by 2', 7' -dichlorodihydrofluorescein diacetate (DCFH-DA) staining. Nuclear translocation of Nrf2 was assessed by immunofluorescence staining and western blotting. Our results indicated that the MTA1 deletion group could differentiate into osteoclasts with larger volume and more TRAP positive. In addition, compared with WT group, KO group cells generated more actin rings. Mechanistically, the loss of MTA1 increased the expression of osteoclast-specific markers, including c-Fos, NFATc1, CTSK and MMP-9. Furthermore, the results of qRT-PCR and western blotting showed that MTA1 deficiency reduced basal Nrf2 expression and inhibited Nrf2-mediated expression of related antioxidant enzymes. Immunofluorescence staining demonstrated that MTA1 deficiency inhibited Nrf2 nuclear translocation. Taken together, the above increased basal and RANKL-induced intracellular ROS levels, leading to enhanced osteoclast formation.
Collapse
Affiliation(s)
- Mingzhe Feng
- Department of Spine SurgeryXi'an Honghui Hospital, School of Medicine, Xi'an Jiaotong UniversityXi'anChina
| | - Lin Liu
- Department of Critical Care MedicineXi'an Honghui Hospital, School of Medicine, Xi'an Jiao Tong UniversityXi'anChina
| | - Zechao Qu
- Department of Spine SurgeryXi'an Honghui Hospital, School of Medicine, Xi'an Jiaotong UniversityXi'anChina
| | - Bo Zhang
- Department of Spine SurgeryXi'an Honghui Hospital, School of Medicine, Xi'an Jiaotong UniversityXi'anChina
| | - Yanjun Wang
- Department of EmergencyXi'an Honghui Hospital, School of Medicine, Xi'an Jiaotong UniversityXi'anChina
| | - Liang Yan
- Department of Spine SurgeryXi'an Honghui Hospital, School of Medicine, Xi'an Jiaotong UniversityXi'anChina
| | - Lingbo Kong
- Department of Spine SurgeryXi'an Honghui Hospital, School of Medicine, Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
4
|
Wu M, Wen L, Zhou Y, Wu W. Role of lncRNA AGAP2-AS1 in Breast Cancer Cell Resistance to Apoptosis by the Regulation of MTA1 Promoter Activity. Technol Cancer Res Treat 2022; 21:15330338221085361. [PMID: 35369814 PMCID: PMC8984859 DOI: 10.1177/15330338221085361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 01/06/2023] Open
Abstract
Introduction Breast cancer (BC) is a common malignant tumor affecting women across the world. LncRNAs are frequently implicated in the course of BC. The current study set out to determine the specific effect of lncRNA AGAP2-AS1 on BC cell resistance to apoptosis. Methods AGAP2-AS1 expression patterns in BC tissues and cells were evaluated. si-AGAP2-AS1 was transfected into MCF-7 cells, followed by the assessment of cell proliferation and apoptosis. In addition to detection of MTA1 expression patterns, the binding relation between AGAP2-AS1 and HuR was verified using RNA pull-down and RNA immunoprecipitation. Next, the regulation enrichment of AGAP2-AS1- and HuR to H3K27ac recruitment in the MTA1 promoter was analyzed. MCF-7 cell resistance to apoptosis was observed after the combined experiment of histone deacetylase inhibitor M344 and si-AGAP2-AS1. Lastly, xenografts tumors were established to detect tumor weight and volume, tumor apoptosis and growth as well as expression of AGAP2-AS1 and MTA1. Results AGAP2-AS1 was overexpressed in BC tissues and cells, and AGAP2-AS1 silencing inhibited cell proliferation but facilitated apoptosis. Physiologically, AGAP2-AS1 bound to HuR to stabilize its own expression, and AGAP2-AS1-HuR complex upregulated H3K27ac levels in the MTA1 promoter region to elevate MTA1 promoter activity and MTA1 expression. H3K27ac upregulation partially-annulled the promotive effect of si-AGAP2-AS1 on BC apoptosis by upregulating MTA1. si-AGAP2-AS1 in vivo inhibited MTA1 expression to enhance apoptosis and suppress tumor growth. Conclusion Collectively, our findings indicated that AGAP2-AS1 bound to HuR to stabilize its own expression, and AGAP2-AS1-HuR complex enhanced H3K27ac levels in the MTA1 promoter region to improve MTA1 promoter activity and MTA1 expression in BC cells, so as to augment BC cell resistance to apoptosis.
Collapse
Affiliation(s)
- Minhua Wu
- Department of thyroid and breast surgery, Ningbo medical center Lihuili
Hospital, Ningbo city, 315040, Zhejiang province, People’s Republic of
China
| | - Limu Wen
- Department of thyroid and breast surgery, Ningbo medical center Lihuili
Hospital, Ningbo city, 315040, Zhejiang province, People’s Republic of
China
| | - Yuxin Zhou
- School of Medicine, Ningbo
University, Ningbo city, 315040, Zhejiang province, People’s Republic of
China
| | - Weizhu Wu
- Department of thyroid and breast surgery, Ningbo medical center Lihuili
Hospital, Ningbo city, 315040, Zhejiang province, People’s Republic of
China
| |
Collapse
|
5
|
Gao J, Liu R, Feng D, Huang W, Huo M, Zhang J, Leng S, Yang Y, Yang T, Yin X, Teng X, Yu H, Yuan B, Wang Y. Snail/PRMT5/NuRD complex contributes to DNA hypermethylation in cervical cancer by TET1 inhibition. Cell Death Differ 2021; 28:2818-2836. [PMID: 33953349 PMCID: PMC8408166 DOI: 10.1038/s41418-021-00786-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/08/2021] [Accepted: 04/15/2021] [Indexed: 02/07/2023] Open
Abstract
The biological function of PRMT5 remains poorly understood in cervical cancer metastasis. Here, we report that PRMT5 physically associates with the transcription factor Snail and the NuRD(MTA1) complex to form a transcriptional-repressive complex that catalyzes the symmetrical histone dimethylation and deacetylation. This study shows that the Snail/PRMT5/NuRD(MTA1) complex targets genes, such as TET1 and E-cadherin, which are critical for epithelial-mesenchymal transition (EMT). This complex also affects the conversion of 5mC to 5hmC. This study demonstrates that the Snail/PRMT5/NuRD(MTA1) complex promotes the invasion and metastasis of cervical cancer in vitro and in vivo. This study also shows that PRMT5 expression is upregulated in cervical cancer and various human cancers, and the PRMT5 inhibitor EPZ015666 suppresses EMT and the invasion potential of cervical cancer cells by disinhibiting the expression of TET1 and increasing 5hmC, suggesting that PRMT5 is a potential target for cancer therapy.
Collapse
Affiliation(s)
- Jie Gao
- grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Inflammatory Biology, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China ,grid.27255.370000 0004 1761 1174The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong China
| | - Ruiqiong Liu
- grid.27255.370000 0004 1761 1174The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong China
| | - Dandan Feng
- grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Inflammatory Biology, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wei Huang
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Cancer Invasion and Metastasis Research, Advanced Innovation Center for Human Brain Protection, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Miaomiao Huo
- grid.506261.60000 0001 0706 7839Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingyao Zhang
- grid.506261.60000 0001 0706 7839Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuai Leng
- grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Inflammatory Biology, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yang Yang
- grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Inflammatory Biology, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tianshu Yang
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Cancer Invasion and Metastasis Research, Advanced Innovation Center for Human Brain Protection, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xin Yin
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Cancer Invasion and Metastasis Research, Advanced Innovation Center for Human Brain Protection, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xu Teng
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Cancer Invasion and Metastasis Research, Advanced Innovation Center for Human Brain Protection, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hefen Yu
- grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Cancer Invasion and Metastasis Research, Advanced Innovation Center for Human Brain Protection, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Baowen Yuan
- grid.506261.60000 0001 0706 7839Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Wang
- grid.265021.20000 0000 9792 1228Tianjin Key Laboratory of Inflammatory Biology, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China ,grid.506261.60000 0001 0706 7839Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Xie X, Wu Q, Zhang K, Liu Y, Zhang N, Chen Q, Wang L, Li W, Zhang J, Liu Y. O-GlcNAc modification regulates MTA1 transcriptional activity during breast cancer cell genotoxic adaptation. Biochim Biophys Acta Gen Subj 2021; 1865:129930. [PMID: 34019948 DOI: 10.1016/j.bbagen.2021.129930] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Chromatin modifier metastasis-associated protein 1 (MTA1), closely associated with tumor angiogenesis in breast cancer, plays an important role in gene expression and cancer cell behavior. Recently, an association between O-GlcNAc transferase (OGT) and MTA1 was identified by mass spectroscopy. However, the potential relationship between MTA1 and O-GlcNAc modification has not yet explored. METHODS In the current study, the role of MTA1 and its O-GlcNAc modification in breast cancer cell genotoxic adaptation was investigated through quantitative proteomics, chromatin immunoprecipitation followed by sequencing (ChIP-seq), transcriptome analysis, and loss- and gain-of-function experiments. RESULTS We demonstrate that the O-GlcNAc modification promotes MTA1 to interaction with chromatin and thus changes the expression of target genes, contributing to breast cancer cell genotoxic adaptation. MTA1 is modified with O-GlcNAc residues at serine (S) residues S237/S241/S246 in adriamycin-adaptive breast cancer cells, and this modification improves the genome-wide interactions of MTA1 with gene promotor regions by enhancing its association with nucleosome remodeling and histone deacetylation (NuRD) complex. Further, O-GlcNAc modification modulates MTA1 chromatin binding, influencing the specific transcriptional regulation of genes involved in the adaptation of breast cancer cells to genotoxic stress. CONCLUSIONS Our findings reveal a previously unrecognized role for O-GlcNAc-modified MTA1 in transcriptional regulation and suggest that the O-GlcNAc modification is a key to the molecular regulation of chemoresistance in breast cancers.
Collapse
Affiliation(s)
- Xueqin Xie
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Qiutong Wu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Keren Zhang
- Clinical Laboratory of BGI Health, BGI-Shenzhen, Shenzhen, China
| | - Yimin Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Nana Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Qiushi Chen
- Clinical Laboratory of BGI Health, BGI-Shenzhen, Shenzhen, China
| | - Lingyan Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Wenli Li
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Jianing Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China..
| | - Yubo Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China..
| |
Collapse
|
7
|
Liu C, Han J, Li X, Huang T, Gao Y, Wang B, Zhang K, Wang S, Zhang W, Li W, Hao Q, Li M, Zhang Y, Zhang C. FOXP3 Inhibits the Metastasis of Breast Cancer by Downregulating the Expression of MTA1. Front Oncol 2021; 11:656190. [PMID: 34307133 PMCID: PMC8293273 DOI: 10.3389/fonc.2021.656190] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/16/2021] [Indexed: 12/09/2022] Open
Abstract
Background FOXP3, as a tumour suppressor gene, has a vital function in inhibiting the metastasis of breast cancer cells, but the mechanisms by which it inhibits metastasis have not been fully elucidated. This study intended to explore a new mechanism by which FOXP3 inhibits breast cancer metastasis. Methods Bioinformatic analysis was performed to identify potential downstream molecules of FOXP3. The function of FOXP3 in inhibiting MTA1 expression at the mRNA and protein levels was verified by real-time PCR and Western blot analysis. The interaction between FOXP3 and the MTA1 promoter was verified by transcriptomic experiments. In vitro and in vivo experiments were used to determine whether the regulation of MTA1 by FOXP3 affected the invasion and migration of breast cancer cells. Immunohistochemistry was adopted to explore the correlation between the expression levels of FOXP3 and MTA1 in breast cancer samples. Results Bioinformatics-based sequencing suggested that MTA1 is a potential downstream molecule of FOXP3. FOXP3 downregulated the expression of MTA1 in breast cancer cells by directly inhibiting MTA1 promoter activity. Importantly, FOXP3’s regulation of MTA1 affected the ability of breast cancer cells to invade and metastasize in vitro and in vivo. Moreover, analysis of clinical specimens showed a significant negative correlation between the expression levels of FOXP3 and MTA1 in breast cancer. Conclusion We systematically explored a new mechanism by which FOXP3 inhibits breast cancer metastasis via the FOXP3-MTA1 pathway.
Collapse
Affiliation(s)
- Chenlin Liu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jun Han
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Xiaoju Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Tonglie Huang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Baolong Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Kuo Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Shuning Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Wangqian Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
8
|
Lin KY, Su TC, Yeh CM, Chao WR, Sung WW. High Expression of MTA1 Predicts Unfavorable Survival in Patients With Oral Squamous Cell Carcinoma. In Vivo 2021; 35:2363-2368. [PMID: 34182519 PMCID: PMC8286510 DOI: 10.21873/invivo.12513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND/AIM Metastasis-associated protein 1 (MTA1) plays a role in ATP-dependent nucleosome disruption activity and histone deacetylase activity and may indicate DNA methylation activity. MTA1 may also be involved in the progression of oral squamous cell carcinoma (OSCC). PATIENTS AND METHODS MTA1 immunoreactivity was analyzed using immunohistochemical (IHC) staining analysis in specimens from 281 OSCC patients. Kaplan-Meier analysis was used to determine the prognostic value of MTA1 for overall survival. RESULTS High MTA1 expression was significantly associated with female gender and lymph node metastasis. Multivariate analyses showed the independent prognostic role of high MTA1 expression in patients with OSCC of poorer mean survival. CONCLUSION MTA1 expression, detected by IHC staining, could be an independent prognostic marker for patients of OSCC.
Collapse
Affiliation(s)
- Kuan-Yu Lin
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
| | - Tzu-Cheng Su
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Chung-Min Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan, R.O.C
| | - Wan-Ru Chao
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- Department of Pathology, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- Department of Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Wen-Wei Sung
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.;
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- Department of Urology, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
| |
Collapse
|
9
|
Kufe DW. MUC1-C in chronic inflammation and carcinogenesis; emergence as a target for cancer treatment. Carcinogenesis 2021; 41:1173-1183. [PMID: 32710608 DOI: 10.1093/carcin/bgaa082] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammation is a highly prevalent consequence of changes in environmental and lifestyle factors that contribute to the development of cancer. The basis for this critical association has largely remained unclear. The MUC1 gene evolved in mammals to protect epithelia from the external environment. The MUC1-C subunit promotes responses found in wound healing and cancer. MUC1-C induces EMT, epigenetic reprogramming, dedifferentiation and pluripotency factor expression, which when prolonged in chronic inflammation promote cancer progression. As discussed in this review, MUC1-C also drives drug resistance and immune evasion, and is an important target for cancer therapeutics now under development.
Collapse
Affiliation(s)
- Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Xu C, Hu Y, Chen B, Li D, Liang R, Shen M, Wu M, Tao M. Metastasis-associated gene 1 (MTA1) enhances cisplatin resistance of malignant pleural mesothelioma by ATR-Chk1-mediated DNA repair. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:670. [PMID: 33987368 PMCID: PMC8106096 DOI: 10.21037/atm-21-941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Malignant pleural mesothelioma (MPM) chemoresistance remains a challenge to oncologists. In our previous study, we demonstrated that the aberrant expression of metastasis-associated gene 1 (MTA1) is associated with carcinogenesis and metastasis in MPM. The aim of the present study was to investigate the mechanism of MTA1 and chemo-resistance in MPM. Methods Western blotting and real-time polymerase chain reaction were used to analyze the protein and mRNA levels. A stable clone with a knockdown of MTA1 was generated with shRNA via lentivirus technology in MPM cell lines. Cell Counting Kit-8 assay and crystal violet assay were used to measure cell viability. Immunochemical staining was employed to detect MTA1 expression in MPM tissues. The cell cycle of MPM cells was determined by phosphohistone H3 staining and flow cytometric analysis. Results The MTA1 protein was upregulated and enhanced cisplatin resistance in MPM. Cisplatin stabilized the expression of the MTA1 protein by inhibiting its ubiquitination, and MTA1 enhanced G2/M cell cycle delay and regulated and protected the tumor genome from chemotherapeutic drugs via participating in the phosphorylation of the ataxia telangiectasia mutated and rad3 related-checkpoint kinase 1 (ATR-Chk1) pathway. Conclusions These data suggest that MTA1 enhances cisplatin resistance by ATR-Chk1-mediated DNA damage repairment and cisplatin stabilizes MTA1 expression via affecting on the ubiquitination pathway of MTA1 in MPM. Our findings indicate that MTA1 could serve as a novel therapeutic target to overcome chemoresistance in MPM.
Collapse
Affiliation(s)
- Caihua Xu
- Department of Oncology of the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yufeng Hu
- Department of Oncology of the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Chen
- Department of Cardiovascular Surgery, Wuxi No. 2 People's Hospital, Wuxi, China
| | - Dapeng Li
- Department of Oncology of the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Rongrui Liang
- Department of Oncology of the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Shen
- Department of Oncology of the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mengyao Wu
- Department of Oncology of the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Tao
- Department of Oncology of the First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| |
Collapse
|
11
|
Dai SL, Wei SS, Zhang C, Li XY, Liu YP, Ma M, Lv HL, Zhang Z, Zhao LM, Shan BE. MTA2 promotes the metastasis of esophageal squamous cell carcinoma via EIF4E-Twist feedback loop. Cancer Sci 2021; 112:1060-1074. [PMID: 33340431 PMCID: PMC7935808 DOI: 10.1111/cas.14778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Metastasis‐associated protein 2 (MTA2) is frequently amplified in many types of cancers; however, the role and underlying molecular mechanism of MTA2 in esophageal squamous cell carcinoma (ESCC) remain unknown. Here, we reported that MTA2 is highly expressed in ESCC tissue and cells, and is closely related to the malignant characteristics and poor prognosis of patients with ESCC. Through in vitro and in vivo experiments, we demonstrated that MTA2 significantly promoted ESCC growth, metastasis, and epithelial‐mesenchymal transition (EMT) progression. This integrative analysis combined with expression microarray showed that MTA2 could interact with eukaryotic initiation factor 4E (EIF4E), which positively regulates the expression of Twist, known as a master regulator of EMT. Moreover, the results of chromatin immunoprecipitation revealed that MTA2 was recruited to the E‐cadherin promoter by Twist, which reduced the acetylation level of the promoter region and thus inhibited expression of E‐cadherin, and subsequently promoted the aggressive progression of ESCC. Collectively, our study provided novel evidence that MTA2 plays an aggressive role in ESCC metastasis by a novel EIF4E‐Twist positive feedback loop, which may provide a potential therapeutic target for the management of ESCC.
Collapse
Affiliation(s)
- Su-Li Dai
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Si-Si Wei
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Cong Zhang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiao-Ya Li
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yue-Ping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ming Ma
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hui-Lai Lv
- Department of Fifth Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenzhen Zhang
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Lian-Mei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bao-En Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
12
|
Li YH, Zhong M, Zang HL, Tian XF. MTA1 Promotes Hepatocellular Carcinoma Progression by Downregulation of DNA-PK-Mediated H1.2 T146 Phosphorylation. Front Oncol 2020; 10:567. [PMID: 32435614 PMCID: PMC7218115 DOI: 10.3389/fonc.2020.00567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/30/2020] [Indexed: 12/24/2022] Open
Abstract
Global incidence and mortality associated with hepatocellular carcinoma (HCC) is steadily increasing. Metastasis-associated 1 (MTA1) can induce tumorigenesis and metastatic progression in HCC. However, the mechanistic details of MTA1-mediated regulation of HCC has not been completely defined. Epigenetic histone modification is closely related to tumor development. Histone cluster 1 H1 family member c (H1.2) is important for epigenetic histone modification and chromatin remodeling; however, whether it has a role in HCC tumorigenesis is not known. In the current study, we confirmed that MTA1 promoted HCC cell growth and migration. Our results further show that MTA1 inhibited the phosphorylation of histone cluster 1 H1 family member c (H1.2) at threonine-146 residue (T146) (H1.2T146ph). MTA1 inhibited H1.2T146ph by mediating proteasomal degradation of the DNA protein kinase (DNA-PK). Pharmacological inhibition of proteasomal degradation of DNA-PK or genetic ablation of E3 ligase mouse double minute 2 (MDM2) rescued expression of DNA-PK, and subsequent phosphorylation of H1.2. MTA1's role in HCC was inhibited by ectopic expression of H1.2T146ph in HCC cell lines. Our results showed that H1.2T146ph can bind to MTA1 target genes. Collectively, our study confirms that MTA1 functions as an oncogene and promotes HCC progression. The epigenetic histone modifier H1.2T146ph exerts critical role in the regulation of MTA1-induced tumorigenesis. MTA1 regulates posttranslational activation of H1.2 by regulating the cognate kinase, DNA-PK, via the ubiquitin proteasome system. MTA1 expression was inversely correlated to both DNA-PK and phosphorylated H1.2 in HCC tissue specimens compared to tumor adjacent normal hepatic tissue, revealing that the MTA1/MDM2/DNA-PK/H1.2 is an important therapeutic axis in HCC.
Collapse
Affiliation(s)
- Yu-Hui Li
- Department of General Surgery, The China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ming Zhong
- Respiratory Department, The China-Japan Union Hospital, Jilin University, Changchun, China
| | - Hong-Liang Zang
- Department of General Surgery, The China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xiao-Feng Tian
- Department of General Surgery, The China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
13
|
Kim MY, Na I, Kim JS, Son SH, Choi S, Lee SE, Kim JH, Jang K, Alterovitz G, Chen Y, van der Vaart A, Won HS, Uversky VN, Kim CG. Rational discovery of antimetastatic agents targeting the intrinsically disordered region of MBD2. SCIENCE ADVANCES 2019; 5:eaav9810. [PMID: 31799386 PMCID: PMC6867884 DOI: 10.1126/sciadv.aav9810] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 09/20/2019] [Indexed: 06/10/2023]
Abstract
Although intrinsically disordered protein regions (IDPRs) are commonly engaged in promiscuous protein-protein interactions (PPIs), using them as drug targets is challenging due to their extreme structural flexibility. We report a rational discovery of inhibitors targeting an IDPR of MBD2 that undergoes disorder-to-order transition upon PPI and is critical for the regulation of the Mi-2/NuRD chromatin remodeling complex (CRC). Computational biology was essential for identifying target site, searching for promising leads, and assessing their binding feasibility and off-target probability. Molecular action of selected leads inhibiting the targeted PPI of MBD2 was validated in vitro and in cell, followed by confirming their inhibitory effects on the epithelial-mesenchymal transition of various cancer cells. Identified lead compounds appeared to potently inhibit cancer metastasis in a murine xenograft tumor model. These results constitute a pioneering example of rationally discovered IDPR-targeting agents and suggest Mi-2/NuRD CRC and/or MBD2 as a promising target for treating cancer metastasis.
Collapse
Affiliation(s)
- Min Young Kim
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Insung Na
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Ji Sook Kim
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
- Department of Pathology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Seung Han Son
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Sungwoo Choi
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Seol Eui Lee
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Ji-Hun Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Korea
| | - Kiseok Jang
- Department of Pathology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Gil Alterovitz
- Boston Children's Hospital/Harvard Medical School, Boston, MA 02115, USA
| | - Yu Chen
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | | | - Hyung-Sik Won
- Department of Biotechnology, Konkuk University, Chungju, Chungbuk 27478, Korea
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Chul Geun Kim
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
14
|
Hata T, Rajabi H, Takahashi H, Yasumizu Y, Li W, Jin C, Long MD, Hu Q, Liu S, Fushimi A, Yamashita N, Kui L, Hong D, Yamamoto M, Miyo M, Hiraki M, Maeda T, Suzuki Y, Samur MK, Kufe D. MUC1-C Activates the NuRD Complex to Drive Dedifferentiation of Triple-Negative Breast Cancer Cells. Cancer Res 2019; 79:5711-5722. [PMID: 31519689 DOI: 10.1158/0008-5472.can-19-1034] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/25/2019] [Accepted: 09/09/2019] [Indexed: 01/04/2023]
Abstract
The NuRD chromatin remodeling and deacetylation complex, which includes MTA1, MBD3, CHD4, and HDAC1 among other components, is of importance for development and cancer progression. The oncogenic mucin 1 (MUC1) C-terminal subunit (MUC1-C) protein activates EZH2 and BMI1 in the epigenetic reprogramming of triple-negative breast cancer (TNBC). However, there is no known link between MUC1-C and chromatin remodeling complexes. Here, we showed that MUC1-C binds directly to the MYC HLH-LZ domain and identified a previously unrecognized MUC1-C→MYC pathway that regulates the NuRD complex. MUC1-C/MYC complexes selectively activated the MTA1 and MBD3 genes and posttranscriptionally induced CHD4 expression in basal- but not luminal-type BC cells. In turn, MUC1-C formed complexes with these NuRD components on the ESR1 promoter. Downregulating MUC1-C decreased MTA1/MBD3/CHD4/HDAC1 occupancy and increased H3K27 acetylation on the ESR1 promoter, with induction of ESR1 expression and downstream estrogen response pathways. Targeting MUC1-C and these NuRD components also induced expression of FOXA1, GATA3, and other markers associated with the luminal phenotype. These findings support a model in which MUC1-C activates the NuRD complex to drive dedifferentiation and reprogramming of TNBC cells. SIGNIFICANCE: MUC1-C directly interacts with MYC to activate the NuRD complex, mediating regulation of the estrogen receptor in triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Tsuyoshi Hata
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hidekazu Takahashi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yota Yasumizu
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Wei Li
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Caining Jin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mark D Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ling Kui
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Deli Hong
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masaaki Yamamoto
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masaaki Miyo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masayuki Hiraki
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Takahiro Maeda
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yozo Suzuki
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mehmet K Samur
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
15
|
Zou G, Bai J, Li D, Chen Y. Effect of metformin on the proliferation, apoptosis, invasion and autophagy of ovarian cancer cells. Exp Ther Med 2019; 18:2086-2094. [PMID: 31452705 PMCID: PMC6704536 DOI: 10.3892/etm.2019.7803] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 04/26/2019] [Indexed: 12/20/2022] Open
Abstract
The present study evaluated the effect of metformin on the SKOV3 ovarian cancer cell line and investigated the underlying mechanism. The inhibitory rate of SKOV3 cells was analyzed by MTT assay. SKOV3 cell apoptosis rate was quantitatively measured using flow cytometry. The effect of metformin on intracellular autophagosomes was observed using electron microscopy. The migration and invasion capabilities of SKOV3 cells were assessed by cell scratch test and Transwell assay. Results demonstrated that. the proliferation rate of SKOV3 cells was significantly inhibited in a time- and concentration-dependent manner following treatment with different concentrations of metformin for 24, 48 and 72 h. The number of migratory cells significantly decreased with increasing concentrations of metformin. The administration of metformin also promoted autophagy of ovarian cancer The expression level of microtubule associated protein 1 light chain 3-α protein was markedly upregulated. The mRNA expression level of metastasis-associated 1 (MTA1) was significantly downregulated following metformin treatment. In conclusion, metformin intervention suppressed SKOV3 proliferation and induced apoptosis in a concentration-dependent manner. Metformin also inhibited the invasion and migration of SKOV3 cells. It was hypothesized that the underlying mechanism of metformin's effect may involve MTA1 downregulation.
Collapse
Affiliation(s)
- Ge Zou
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Jie Bai
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Dandan Li
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yan Chen
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
16
|
Guo N, Shen G, Zhang Y, Moustafa AA, Ge D, You Z. Interleukin-17 Promotes Migration and Invasion of Human Cancer Cells Through Upregulation of MTA1 Expression. Front Oncol 2019; 9:546. [PMID: 31281798 PMCID: PMC6596356 DOI: 10.3389/fonc.2019.00546] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/04/2019] [Indexed: 12/21/2022] Open
Abstract
Interleukin-17 (IL-17) has been shown to promote development of prostate, colon, skin, lung, breast, and pancreatic cancer. The purpose of this study was to determine if IL-17 regulates MTA1 expression and its biological consequences. Human cervical cancer HeLa and human prostate cancer DU-145 cell lines were used to test if IL-17 regulates metastasis associated 1 (MTA1) mRNA and protein expression using quantitative reverse transcription-polymerase chain reaction and Western blot analysis, respectively. Cell migration and invasion were studied using wound healing assays and invasion chamber assays. Thirty-four human cervical tissues were stained for IL-17 and MTA1 using immunohistochemical staining. We found that IL-17 increased MTA1 mRNA and protein expression in both cell lines. Cell migration was accelerated by IL-17, which was abolished by knockdown of MTA1 expression with small interference RNA (siRNA). Further, cell invasion was enhanced by IL-17, which was eliminated by MTA1 knockdown. Human cervical intra-epithelial neoplasia (CIN) and cervical cancer tissues had increased number of IL-17-positive cells and MTA1 expression compared to normal cervical tissues. The number of IL-17-positive cells was positively correlated with MTA1 expression. These findings demonstrate that IL-17 upregulates MTA1 mRNA and protein expression to promote HeLa and DU-145 cell migration and invasion.
Collapse
Affiliation(s)
- Na Guo
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA, United States.,Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ge Shen
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA, United States
| | - Ying Zhang
- Department of Gynecology, Guangyuan First People's Hospital, Guangyuan, China
| | - Ahmed A Moustafa
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA, United States
| | - Dongxia Ge
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA, United States
| | - Zongbing You
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA, United States.,Department of Orthopaedic Surgery, Tulane University, New Orleans, LA, United States.,Tulane Cancer Center and Louisiana Cancer Research Consortium, Tulane University, New Orleans, LA, United States.,Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, LA, United States.,Tulane Center for Aging, Tulane University, New Orleans, LA, United States.,Southeast Louisiana Veterans Health Care System, New Orleans, LA, United States
| |
Collapse
|
17
|
Abstract
Cancer-initiating cells (CIC) are the driving force in tumor progression. There is strong evidence that CIC fulfill this task via exosomes (TEX), which modulate and reprogram stroma, nontransformed cells, and non-CIC. Characterization of CIC, besides others, builds on expression of CIC markers, many of which are known as metastasis-associated molecules. We here discuss that the linkage between CIC/CIC-TEX and metastasis-associated molecules is not fortuitously, but relies on the contribution of these markers to TEX biogenesis including loading and TEX target interactions. In addition, CIC markers contribute to TEX binding- and uptake-promoted activation of signaling cascades, transcription initiation, and translational control. Our point of view will be outlined for pancreas and colon CIC highly expressing CD44v6, Tspan8, EPCAM, claudin7, and LGR5, which distinctly but coordinately contribute to tumor progression. Despite overwhelming progress in unraveling the metastatic cascade and the multiple tasks taken over by CIC-TEX, there remains a considerable gap in linking CIC biomarkers, TEX, and TEX-initiated target modulation with metastasis. We will try to outline possible bridges, which could allow depicting pathways for new and expectedly powerful therapeutic interference with tumor progression.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - Margot Zöller
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany.
| |
Collapse
|
18
|
Wu Y, Zhai W, Zhou X, Wang Z, Lin Y, Ran L, Qi Y, Gao Y. HLA-A2-Restricted Epitopes Identified from MTA1 Could Elicit Antigen-Specific Cytotoxic T Lymphocyte Response. J Immunol Res 2018; 2018:2942679. [PMID: 30596107 PMCID: PMC6286779 DOI: 10.1155/2018/2942679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/31/2018] [Accepted: 09/13/2018] [Indexed: 02/06/2023] Open
Abstract
Overexpression of metastasis-associated protein 1 (MTA1) has been observed in many human malignancies and is significantly related to tumor invasion and metastasis, therapeutic resistance to radiation and chemotherapy, making MTA1 an ideal candidate tumor antigen. We identified several human leukocyte antigen- (HLA-) A2-restricted epitopes in MTA1 and evaluated their binding ability to HLA-A∗0201 molecules. Subsequently, a recombinant fragment encompassing the dominant epitopes, MTA1(1-283), was expressed, and the abilities of the selected epitopes of MTA1 and the MTA1(1-283) fragment to induce cytotoxic T lymphocytes (CTLs) were examined. Our results indicated that the epitopes and MTA1(1-283) fragment elicited HLA-A2-restricted and antigen-specific CTL responses both in vitro and in vivo. The new epitopes identified here may help promote the development of new therapeutic vaccines for HLA-A2+ patients expressing MTA1.
Collapse
Affiliation(s)
- Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiuman Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhiwei Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Lin
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ling Ran
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
19
|
Wang S, Li Q, Wang Y, Li X, Wang R, Kang Y, Xue X, Meng R, Wei Q, Feng X. Upregulation of circ-UBAP2 predicts poor prognosis and promotes triple-negative breast cancer progression through the miR-661/MTA1 pathway. Biochem Biophys Res Commun 2018; 505:996-1002. [PMID: 30314706 DOI: 10.1016/j.bbrc.2018.10.026] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/05/2018] [Indexed: 02/06/2023]
Abstract
Recently, a large number of studies have shown that circular RNA (circRNA) is involved in the development and progression of human cancer. However, its role in triple-negative breast cancer (TNBC) remains largely unknown. In this study, a novel circRNA, circ-UBAP2 (hsa_circ_0001846), was shown to be markedly upregulated in TNBC. Moreover, high circ-UBAP2 expression was closely associated with larger tumour size (p = 0.003), advanced TNM stage (p = 0.005), lymph node metastasis (p = 0.002), and unfavourable prognosis (p = 0.0256). Functionally, lentivirus-mediated stable knockdown of circ-UBAP2 dramatically weakened the ability of TNBC cells to proliferate and migrate and induced apoptosis in vitro and in vivo. Regarding the mechanism, we found that circ-UBAP2 was mainly observed in the cytoplasm and was capable of sponging miRNA-661 to increase the expression of the oncogene MTA1. Additionally, silencing of miRNA-661 or overexpression of MTA1 could partially rescue the attenuated malignant phenotype caused by circ-UBAP2 knockdown. Taken together, our data reveal that circ-UBAP2 plays a vital regulatory role in TNBC via the miR-661/MTA1 axis and may serve as a promising therapeutic target for TNBC patients.
Collapse
Affiliation(s)
- Shengting Wang
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China
| | - Qian Li
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China
| | - Yufang Wang
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China
| | - Xiaoming Li
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China
| | - Rui Wang
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China
| | - Yuhua Kang
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China
| | - Xukai Xue
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, 710054, China
| | - Rui Meng
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China
| | - Qi Wei
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China
| | - Xinghua Feng
- Department of Clinical Medicine, Peihua University, Xi'an, 710125, China.
| |
Collapse
|
20
|
Yu R, Cai L, Chi Y, Ding X, Wu X. miR‑377 targets CUL4A and regulates metastatic capability in ovarian cancer. Int J Mol Med 2018; 41:3147-3156. [PMID: 29512715 PMCID: PMC5881808 DOI: 10.3892/ijmm.2018.3540] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 01/31/2018] [Indexed: 12/20/2022] Open
Abstract
The incidence and recurrence rates of ovarian cancer are still high, and once the disease metastasizes, it is nearly always fatal. Cullin 4A (CUL4A) serves a significant role in tumourigenesis and tumour progression; however, the effect and mechanisms underlying CUL4A overexpression are still unknown. The role of microRNAs (miRs) in the regulation of metastatic capability in ovarian cancer cell lines was investigated. The interaction between miR‑377 and CUL4A was investigated using bioinformatics analyses and dual‑luciferase reporter assays. Furthermore, miR‑377 mRNA and protein levels were detected using reverse transcription‑quantitative polymerase chain reaction and western blotting, respectively and cell migration and invasion were detected using a Transwell assay. Results revealed that CUL4A expression was negatively associated with miR‑377 levels in ovarian cancer tissues and cell lines. Through in silico analysis, the targeting effect of miR‑377 on CUL4A was verified. Ectopic expression of miR‑377 in SKOV3 cells downregulated the level of CUL4A, and significantly reduced the migratory ability of the cells. miR‑377 overexpression led to reduced activity of the Wnt/β‑catenin signaling pathway, and regulated the expression of matrix metalloproteinase‑2, and 9, and epithelial‑mesenchymal transition (EMT)‑associated protein. These results suggested that miR‑377 is a significant negative regulator of CUL4A that controls cancer cell progression in ovarian cancer cell lines.
Collapse
Affiliation(s)
- Rufen Yu
- Department of Obstetrics and Gynecology, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P. R. China
| | - Limei Cai
- Department of Obstetrics and Gynecology, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P. R. China
| | - Yingui Chi
- Department of Obstetrics and Gynecology, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P. R. China
| | - Xiangcui Ding
- Department of Obstetrics and Gynecology, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P. R. China
| | - Xueqing Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, The First Provincial Wenzhou Hospital of Zhejiang, Wenzhou, Zhejiang 325000, P. R. China
| |
Collapse
|
21
|
Understanding the Progression of Bone Metastases to Identify Novel Therapeutic Targets. Int J Mol Sci 2018; 19:ijms19010148. [PMID: 29300334 PMCID: PMC5796097 DOI: 10.3390/ijms19010148] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/22/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022] Open
Abstract
Bone is one of the most preferential target site for cancer metastases, particularly for prostate, breast, kidney, lung and thyroid primary tumours. Indeed, numerous chemical signals and growth factors produced by the bone microenvironment constitute factors promoting cancer cell invasion and aggression. After reviewing the different theories proposed to provide mechanism for metastatic progression, we report on the gene expression profile of bone-seeking cancer cells. We also discuss the cross-talk between the bone microenvironment and invading cells, which impacts on the tumour actions on surrounding bone tissue. Lastly, we detail therapies for bone metastases. Due to poor prognosis for patients, the strategies mainly aim at reducing the impact of skeletal-related events on patients' quality of life. However, recent advances have led to a better understanding of molecular mechanisms underlying bone metastases progression, and therefore of novel therapeutic targets.
Collapse
|