1
|
He W, Zhang J, Wen S, Li Y, Shen L, Zhou T, Wen Q, Fan Y. Epigenetic identification of LTBP4 as a putative tumor suppressor in breast cancer. Epigenomics 2024; 16:999-1012. [PMID: 39193795 PMCID: PMC11404579 DOI: 10.1080/17501911.2024.2388017] [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: 04/06/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Aim: To explore the LTBP4's expression, prognostic significance and molecular mechanism of action in breast cancer (BC).Methods: On the basis of omics datasets and experiments, we conducted a synthetical analysis of LTBP4 in BC.Results & conclusion: LTBP4 was downregulated in BC with high promoter methylation and low genetic alteration. DNA methylation was negatively associated with LTBP4 mRNA expression. Higher LTBP4 associated with better survival. LTBP4 was enrichment in extracellular matrix receptor interactions, cell adhesion molecules, cell cycle and MAPK pathway. LTBP4 expression and methylation were positively and negatively associated with tumor infiltrating immune cells, respectively. In conclusion, LTBP4 is a putative tumor suppressor in BC, which expression is regulated by DNA methylation and relates with prognosis.
Collapse
Affiliation(s)
- Wenfeng He
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, 646000, Luzhou, PR China
| | - Jingheng Zhang
- Department of Oncology, The People’s Hospital of Luzhou, 646000, Luzhou, PR China
| | - Siyuan Wen
- Faculty of Clinical Medicine, Southwest Medical University, 646000, Luzhou, PR China
| | - Ying Li
- Faculty of Clinical Medicine, Southwest Medical University, 646000, Luzhou, PR China
| | - Lin Shen
- Faculty of Clinical Medicine, Southwest Medical University, 646000, Luzhou, PR China
| | - Tiejun Zhou
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, 646000, Luzhou, PR China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, 646000, Luzhou, PR China
| | - Yu Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, 646000, Luzhou, PR China
| |
Collapse
|
2
|
Berkel C, Cacan E. The expression of O-linked glycosyltransferase GALNT7 in breast cancer is dependent on estrogen-, progesterone-, and HER2-receptor status, with prognostic implications. Glycoconj J 2023; 40:631-644. [PMID: 37947928 DOI: 10.1007/s10719-023-10137-4] [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: 04/04/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
GALNT7 is a glycosyltransferase enzyme transferring N-acetylgalactosamine to initiate O-linked glycosylation in the Golgi apparatus. Breast cancer is the most common cancer in women globally. Estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2; ERBB2) are important biomarkers in the prognosis and molecular subtyping of breast cancer. Here, we showed that ER-positive, PR-positive or HER2-positive breast tumors have higher expression of GALNT7 compared to ER-negative, PR-negative or HER2-negative breast tumors, respectively. We found that CpG-aggregated methylation of GALNT7 gene is decreased, and in parallel, its transcript levels are increased in breast cancer compared to healthy breast tissue. We observed that the difference in the expression of GALNT7 between negative and positive status of the receptors is the highest for HER2, followed by ER and PR, pointing that HER2 might be relatively more influential than ER and PR on the expression of GALNT7 in breast cancer. We reported that basal-like breast tumors have decreased expression of GALNT7 compared to non-basal-like tumors, and that high GALNT7 expression is associated with favorable relapse-free and distant metastasis-free survival in HER2 status-dependent manner in breast cancer patients. Moreover, we showed that GALNT7 expression in breast cancer is cell type- (epithelial vs stromal cells), tumor grade- and ethnicity-dependent. Combined, we propose that GALNT7 might contribute to different clinical outcomes depending on the receptor status in breast cancer, and that a better understanding of GALNT7 and its function in the context of breast cancer is needed.
Collapse
Affiliation(s)
- Caglar Berkel
- Department of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Turkey.
| | - Ercan Cacan
- Department of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Turkey
| |
Collapse
|
3
|
Bai Y, Yuan F, Yu J, Si Y, Zheng Y, Li D. A BIRC5 High COD1 Low Cancer Tissue Phenotype Indicates Poorer Prognosis of Metastatic Breast Cancer Patients. Cancer Inform 2022; 21:11769351221096655. [PMID: 35734521 PMCID: PMC9208035 DOI: 10.1177/11769351221096655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Extensive data research is helpful to find sensitive biomarkers for prognostic prediction of metastatic breast cancer. Through analyzing multiple GEO datasets, literature retrieval, and verified in GEPIA datasets, we identify BIRC5 (Baculoviral IAP repeat containing 5) and CDO1 (Cysteine dioxygenase type 1) as DEGs (differentially expressed genes) between breast tumor and normal tissue and DEGs between metastatic breast cancer and breast cancer in situ. Then, we performed a series of in silico studies on BIRC5 and CDO1 using online tools including the UALCAN, TIMER, TCGA-BRCA, LinkedOmics Kaplan-Meier Plotter, and an R script for analysis. To verify the association of 2 genes expression and patients’ clinical data, we detected BIRC5 and CDO1 mRNA in the tissue of 48 breast cancer patients. The results showed the tumor with BIRC5high CDO1low expression generally indicated patients’ shorter overall (OS) and relapse-free survival (RFS). Specifically, BIRC5 and CDO1 levels significantly affect OS or RFS in patients with Lymph node metastasis and molecular subtypes of TNBC (triple-negative breast cancer) and Luminal A. A BIRC5high tumor displayed a purer tumor purity and expressed more KIR receptors on NK cells while activating more FOXP3+CD25+ Treg cells. The CDO1low tumors infiltrated with more immunocytes leading to less tumor purity. In our verified experiment, BIRC5 mRNA level in patients with stage III and over was significantly higher than in patients with stage 0 to II, but there were no significant differences among molecular subtyping groups; TNBC tissue expressed lower CDO1 mRNA level than HER2+ and Luminal type cancer tissue. In conclusion, a BIRC5high CDO1low expression type in breast cancer tissue indicates a poorer prognosis of patients. The potential mechanism might be increased BIRC5 expression in cancer tissue is likely to accompany NK cells inhibition, activating more Treg cells, and lacking effective CD8+ T cells proliferation. Meanwhile, CDO1 level is positively related to more immunocytes infiltration.
Collapse
Affiliation(s)
- Yujie Bai
- Department of Microbiology, School of Basic Medical of Science, Wuhan University, Wuhan, China.,Department of Scientific Research and Education, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Feng Yuan
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan, China
| | - Jing Yu
- Department of Blood Transfusion, Wuhan No.1 Hospital/Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yibei Si
- Department of Microbiology, School of Basic Medical of Science, Wuhan University, Wuhan, China
| | - Yiwen Zheng
- Department of Microbiology, School of Basic Medical of Science, Wuhan University, Wuhan, China
| | - Dongqing Li
- Department of Microbiology, School of Basic Medical of Science, Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Fang Y, Liu J, Zhang Q, She C, Zheng R, Zhang R, Chen Z, Chen C, Wu J. Overexpressed VDAC1 in breast cancer as a novel prognostic biomarker and correlates with immune infiltrates. World J Surg Oncol 2022; 20:211. [PMID: 35729567 PMCID: PMC9215028 DOI: 10.1186/s12957-022-02667-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/28/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND More and more evidence suggests that cancer is a mitochondrial metabolic disease recently and mitochondria dysfunction is critical to tumorigenesis. As a gatekeeper of mitochondria, the voltage-dependent anion channel 1 (VDAC1) is associated with the development of breast cancer (BC). However, its potential mechanism and clinical significance remain unclear; thus, in this research, we aimed to explore it. METHODS VDAC1 expression in BC tissues and normal tissues was obtained from The Cancer Genome Atlas (TCGA) and validated by datasets from the gene expression omnibus (GEO) database. Then, the relationships between VDAC1 expression and clinicopathological features were analyzed. Receiver operating characteristics (ROC) curves were used to identify the diagnostic value of VDAC1. The prognostic value was evaluated by Kaplan-Meier survival curves and Cox regression analysis. VDAC1 with its co-expression genes were subjected to enrichment analysis to explore potential mechanisms in BC and the protein-protein interaction (PPI) network was constructed. At last, the association between VDAC1 expression and infiltration levels of immune cell infiltration by various methods, as well as their corresponding markers, was analyzed. We also analyzed the correction between VDAC1 expression and eight immune checkpoint genes and the tumor immune dysfunction and exclusion (TIDE) scores of each BC sample in TCGA were calculated and the differences between high and low VDAC1 expression groups were analyzed. RESULTS VDAC1 expression was remarkably elevated in BC (p < 0.001), and high expression of VDAC1 was associated with the positive expression of ER (p = 0.004), PR (p = 0.033), and HER2 (p = 0.001). ROC analysis suggested that VDAC1 had diagnosed value in BC. The Kaplan-Meier analysis suggested that higher expression of VDAC1 was associated with shorter overall survival (OS), and further Cox regression analysis revealed that VDAC1 was an independent factor of unfavorable prognosis in BC patients. Enrichment analysis of VDAC1 and its co-expression suggested that VDAC1 was related to the regulation of mitochondrial energy metabolism and protein modification, and the HIF-1 singing pathway might be the potential mechanism in BC. Notably, we found that VDAC1 expression was infiltration levels of most types of immune cells, as well as the expression of marker genes of immune cells. The ICGs PDCD1, CTLA4, LAG3, SIGLEC15, and TIGIT were negatively corrected with VDAC1 expression in BC. TIDE scores between the low and high expression groups showed no difference. CONCLUSION Overexpressed VDAC1 in BC could be severed as a novel biomarker for diagnosis and VDAC1 was an independent factor for adverse prognosis prediction. Our study revealed that VDAC1 might inhibit tumor immunity and might be a novel therapeutic target in BC.
Collapse
Affiliation(s)
- Yutong Fang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Junpeng Liu
- Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Qunchen Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Chuanghong She
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Rongji Zheng
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Rendong Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Zexiao Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Chunfa Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Jundong Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| |
Collapse
|
5
|
Jang HN, Moon SJ, Jung KC, Kim SW, Kim H, Han D, Kim JH. Mass Spectrometry-Based Proteomic Discovery of Prognostic Biomarkers in Adrenal Cortical Carcinoma. Cancers (Basel) 2021; 13:3890. [PMID: 34359790 PMCID: PMC8345732 DOI: 10.3390/cancers13153890] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/30/2022] Open
Abstract
Adrenal cortical carcinoma (ACC) is an extremely rare disease with a variable prognosis. Current prognostic markers have limitations in identifying patients with a poor prognosis. Herein, we aimed to investigate the prognostic protein biomarkers of ACC using mass-spectrometry-based proteomics. We performed the liquid chromatography-tandem mass spectrometry (LC-MS/MS) using formalin-fixed paraffin-embedded (FFPE) tissues of 45 adrenal tumors. Then, we selected 117 differentially expressed proteins (DEPs) among tumors with different stages using the machine learning algorithm. Next, we conducted a survival analysis to assess whether the levels of DEPs were related to survival. Among 117 DEPs, HNRNPA1, C8A, CHMP6, LTBP4, SPR, NCEH1, MRPS23, POLDIP2, and WBSCR16 were significantly correlated with the survival of ACC. In age- and stage-adjusted Cox proportional hazard regression models, only HNRNPA1, LTBP4, MRPS23, POLDIP2, and WBSCR16 expression remained significant. These five proteins were also validated in TCGA data as the prognostic biomarkers. In this study, we found that HNRNPA1, LTBP4, MRPS23, POLDIP2, and WBSCR16 were protein biomarkers for predicting the prognosis of ACC.
Collapse
Affiliation(s)
- Han Na Jang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (H.N.J.); (S.J.M.); (S.W.K.)
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Sun Joon Moon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (H.N.J.); (S.J.M.); (S.W.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03080, Korea
| | - Kyeong Cheon Jung
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea;
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Sang Wan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (H.N.J.); (S.J.M.); (S.W.K.)
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul 03080, Korea
| | - Hyeyoon Kim
- Proteomics Core Facility, Biomedical Research Institute Seoul National University Hospital, Seoul 03080, Korea;
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute Seoul National University Hospital, Seoul 03080, Korea;
| | - Jung Hee Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (H.N.J.); (S.J.M.); (S.W.K.)
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
6
|
Su CT, Urban Z. LTBP4 in Health and Disease. Genes (Basel) 2021; 12:genes12060795. [PMID: 34071145 PMCID: PMC8224675 DOI: 10.3390/genes12060795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/14/2021] [Accepted: 05/21/2021] [Indexed: 12/20/2022] Open
Abstract
Latent transforming growth factor β (TGFβ)-binding protein (LTBP) 4, a member of the LTBP family, shows structural homology with fibrillins. Both these protein types are characterized by calcium-binding epidermal growth factor-like repeats interspersed with 8-cysteine domains. Based on its domain composition and distribution, LTBP4 is thought to adopt an extended structure, facilitating the linear deposition of tropoelastin onto microfibrils. In humans, mutations in LTBP4 result in autosomal recessive cutis laxa type 1C, characterized by redundant skin, pulmonary emphysema, and valvular heart disease. LTBP4 is an essential regulator of TGFβ signaling and is related to development, immunity, injury repair, and diseases, playing a central role in regulating inflammation, fibrosis, and cancer progression. In this review, we focus on medical disorders or diseases that may be manipulated by LTBP4 in order to enhance the understanding of this protein.
Collapse
Affiliation(s)
- Chi-Ting Su
- Department of Internal Medicine, Renal Division, National Taiwan University Hospital Yunlin Branch, Douliu 640, Taiwan;
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Medicine, National Taiwan University Cancer Center Hospital, Taipei 106, Taiwan
| | - Zsolt Urban
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence: ; Tel.: +1-412-648-8269
| |
Collapse
|
7
|
He B, Chen J, Song W, Bai Y. miR-646/TET1 mediated demethylation of IRX1 promoter upregulates HIST2H2BE and promotes the progression of invasive ductal carcinoma. Genomics 2021; 113:1469-1481. [PMID: 33667646 DOI: 10.1016/j.ygeno.2020.12.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND This study aimed to explore role of miR-646 in breast IDC. METHODS miR-646, TET1, IRX1, and HIST2H2BE expression was detected by RT-qPCR and/or Western blot analysis. The methylation status of IRX1 promoter region was evaluated by methylation specific PCR. ChIP assay was used to determine the enrichment of TET1 at IRX1 promoter region. Loss- and gain-of functions were performed to determine the roles of miR-646, TET1, IRX1, and HIST2H2BE in cell proliferation, migration, invasion, and apoptosis. The tumor growth, volume, weight, and apoptosis status were measured. RESULTS miR-646 was upregulated while TET1 was downregulated in IDC tissues. miR-646 targeted TET1. Downregulated TET1 impairs demethylation of IRX1 promoter region resulting in reduced expression of IRX1, which subsequently leads to upregulation of HIST2H2BE in IDC. Consequently, elevated HIST2H2BE promotes progression of IDC. CONCLUSION Our study has demonstrated that miR-646 facilitates the tumorigenesis of IDC via regulating TET1/IRX1/HIST2H2BE axis.
Collapse
Affiliation(s)
- Baoxia He
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450003, PR China.
| | - Jinhua Chen
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450003, PR China
| | - Wenping Song
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450003, PR China
| | - Yongtao Bai
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450003, PR China
| |
Collapse
|
8
|
Cai M, Li H, Chen R, Zhou X. MRPL13 Promotes Tumor Cell Proliferation, Migration and EMT Process in Breast Cancer Through the PI3K-AKT-mTOR Pathway. Cancer Manag Res 2021; 13:2009-2024. [PMID: 33658859 PMCID: PMC7920513 DOI: 10.2147/cmar.s296038] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose Breast cancer (BC), with varying histopathology, biology and response to systemic treatment, is the second leading cause of cancer-related mortality. Previous studies have inferred that the expression of mitochondrial ribosomal proteins (MRPs) is possibly related to the occurrence/progression of BC. MRPL13 might be one of the potential MRP candidates that are involved in BC tumorigenesis, but its role in BC has rarely been reported. The purpose of the current study was to evaluate the prognostic significance of MRPL13, as well as to explore its potential biological functions in BC. Materials and Methods A series of bioinformatic and statistical methods were adopted to assess the MRPL13 expression profile, its relationship with clinicopathological characteristics, copy number variation (CNV), impact on clinical outcomes and relevant functions. All the results are analysed by 1097 BC patients collected from The Cancer Genome Atlas (TCGA) dataset and 52 clinical samples for immunohistochemistry (IHC) assay. Results The results demonstrated that the expression of MRPL13 in BC tissues was remarkably elevated than that in normal breast tissues. In addition, the Kaplan-Meier curves and Cox model indicated that patients with high MRPL13 expression were connected to a worse prognosis, heralding the independent prognostic value of this protein in BC. Moreover, an enrichment analysis showed that MRPL13 was mainly involved in cell cycle/division-related, RNA processing (degradation/splicing), MYC targets and the MTORC1 pathways. In addition, RNA interference (RNAi)-mediated MRPL13 silencing remarkedly inhibited proliferation and migration as well as the expression of EMT-related genes of BC cells in vitro. Mechanistically, attenuation of MRPL13 significantly suppressed the phosphorylation of AKT and mTOR, which could be partially abolished by 740Y-P (a PI3K agonist). Conclusion Our results provide evidence for the first time that increased MRPL13 expression correlates with adverse clinicopathological variables and unfavorable clinical outcomes of BC patients. Knockdown of MRPL13 restrains the proliferation and migration potential and EMT process of BC through inhibiting PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Miaomiao Cai
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, People's Republic of China
| | - Hanning Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Runfa Chen
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, People's Republic of China
| | - Xiang Zhou
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, People's Republic of China
| |
Collapse
|
9
|
Cai R, Wang P, Zhao X, Lu X, Deng R, Wang X, Su Z, Hong C, Lin J. LTBP1 promotes esophageal squamous cell carcinoma progression through epithelial-mesenchymal transition and cancer-associated fibroblasts transformation. J Transl Med 2020; 18:139. [PMID: 32216815 PMCID: PMC7098101 DOI: 10.1186/s12967-020-02310-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent cancers worldwide. Due to its high morbidity and mortality rates, it is urgent to find a molecular target that contributes to esophageal carcinogenesis and progression. In this research, we aimed to investigate the functions of Latent transforming growth factor β binding protein 1(LTBP1) in ESCC progression and elucidate the underlying mechanisms. METHODS The tandem mass tag-based quantitative proteomic approach was applied to screen the differentially expressed proteins (DEPs) between 3 cases of ESCC tumor samples and paired normal tissues. Then the DEPs were validated in human ESCC tissues using western blot assays and GEPIA database respectively. The expression level of LTBP1 was detected in 152 cases of ESCC tissues and paired normal tissues. Loss-of-function assays were performed to detect the function of LTBP1 in vivo and in vitro. Immunofluorescence and Western blot assays were used to detect the expression of apoptosis, epithelial-mesenchymal transition (EMT) and cancer-associated fibroblasts (CAFs) markers. RESULTS A total of 39 proteins were screened to be up-regulated (ratio > 2.0) in all three ESCC tissues. The results of immunohistochemistry assays indicated that the expression level of LTBP1 was higher in ESCC tissues than that in paired normal tissues (p < 0.001). Overexpression of LTBP1 was positively associated with lymphatic metastasis in ESCC (p = 0.002). Down-regulation of LTBP1 inhibited the invasion and migration as well as metastatic abilities in vitro and in vivo. It was also observed the down-regulation of LTBP1 not only decreased the mesenchymal phenotypes but also inhibited TGFβ-induced EMT in ESCC cells. We further found that down-regulation of LTBP1 enhanced ESCC cells' sensitivity to 5-FU treatment. Inhibition of LTBP1 expression could also attenuate induction of CAFs transformation and restrain fibroblast express fibronectin (FN1) in ESCC cells. CONCLUSION Overexpression of LTBP1 was associated with lymph node metastasis in ESCC. Our results indicated that LTBP1 not only increased the malignant behaviors of ESCC cells but also induced EMT and CAFs transformation. Our studies suggested an oncogenic role of LTBP1 in ESCC progression and it may serve as a potential therapeutic target for ESCC patients.
Collapse
Affiliation(s)
- Rui Cai
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Ping Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Xin Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Xiansheng Lu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Ruxia Deng
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Xiaoyu Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Zhaoji Su
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Chang Hong
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China. .,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China.
| |
Collapse
|
10
|
Song G, He L, Yang X, Yang Y, Cai X, Liu K, Feng G. Identification of aberrant gene expression during breast ductal carcinoma in situ progression to invasive ductal carcinoma. J Int Med Res 2019; 48:300060518815364. [PMID: 30712460 PMCID: PMC7140215 DOI: 10.1177/0300060518815364] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Guiqin Song
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, the Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, P.R. China.,Department of Biology, North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Lang He
- Department of Oncology, the Fifth People's Hospital of Chengdu, The Second Clinical Medical School of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Xiaolin Yang
- Department of Biology, North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Yan Yang
- Sichuan Chidingshengtong Biotechnology Co., Ltd., Chengdu, Sichuan, P.R. China
| | - Xiaoming Cai
- Department of Biology, North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, the Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, P.R. China.,Precision Medicine Center, Nanchong Central Hospital, Nanchong, Sichuan, P.R. China
| | - Gang Feng
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, the Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, P.R. China.,Precision Medicine Center, Nanchong Central Hospital, Nanchong, Sichuan, P.R. China
| |
Collapse
|
11
|
Qiu L, Wang T, Xu X, Wu Y, Tang Q, Chen K. Long Non-Coding RNAs in Hepatitis B Virus-Related Hepatocellular Carcinoma: Regulation, Functions, and Underlying Mechanisms. Int J Mol Sci 2017; 18:ijms18122505. [PMID: 29168767 PMCID: PMC5751108 DOI: 10.3390/ijms18122505] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/13/2017] [Accepted: 11/20/2017] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of cancer death in the world. Hepatitis B virus (HBV) and its X gene-encoded protein (HBx) play important roles in the progression of HCC. Although long non-coding RNAs (lncRNAs) cannot encode proteins, growing evidence indicates that they play essential roles in HCC progression, and contribute to cell proliferation, invasion and metastasis, autophagy, and apoptosis by targeting a large number of pivotal protein-coding genes, miRNAs, and signaling pathways. In this review, we briefly outline recent findings of differentially expressed lncRNAs in HBV-related HCC, with particular focus on several key lncRNAs, and discuss their regulation by HBV/HBx, their functions, and their underlying molecular mechanisms in the progression of HCC.
Collapse
Affiliation(s)
- Lipeng Qiu
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| | - Tao Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| | - Xiuquan Xu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China.
| | - Yihang Wu
- Department of Pharmacy, College of Life Sciences, China Jiliang University, Hangzhou 310018, China.
| | - Qi Tang
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| | - Keping Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
12
|
Lu J, Liu Q, Wang L, Tu W, Chu H, Ding W, Jiang S, Ma Y, Shi X, Pu W, Zhou X, Jin L, Wang J, Wu W. Increased expression of latent TGF-β-binding protein 4 affects the fibrotic process in scleroderma by TGF-β/SMAD signaling. J Transl Med 2017; 97:591-601. [PMID: 28263294 DOI: 10.1038/labinvest.2017.20] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/03/2017] [Accepted: 01/13/2017] [Indexed: 01/06/2023] Open
Abstract
Scleroderma is a fibrosis-related disorder characterized by cutaneous and internal organ fibrosis, and excessive collagen deposition in extracellular matrix (ECM) is a major cause of fibrosis. Transforming growth factor-β (TGF-β)/SMAD signaling has a central role in the pathogenesis of fibrosis by inducing abnormal collagen accumulation in ECM, and latent TGF-β-binding protein 4 (LTBP-4) affects the secretion of latent TGF-β to ECM. A previous study indicated that bleomycin (BLM) treatment increased LTBP-4 expression in lung fibroblasts of Thy-1 knockout mice with lung fibrosis, and LTBP-4 further promoted TGF-β bioavailability as well as SMAD3 phosphorylation. However, the expression and function of LTBP-4 in human scleroderma remain unclear. We aimed to investigate the potential role of LTBP-4 in scleroderma through clinical, in vivo and in vitro studies. LTBP-4 and TGF-β expressions were significantly upregulated in systemic scleroderma (SSc) patients' plasma compared with normal controls (LTBP-4, 1,215±100.2 vs 542.8±41.7 ng/ml, P<0.0001; TGF-β, 1.5±0.2 vs 0.7±0.1 ng/ml, P=0.0031), while no significant difference was found between localized scleroderma (LSc) and normal controls. The plasma concentrations of LTBP-4 and TGF-β were even higher in SSc patients with lung fibrosis (LTBP-4, 1462± 137.3 vs 892.8±113.4 ng/ml, P=0.0037; TGF-β, 2.0±0.4 vs 0.9±0.2 ng/ml, P=0.0212) and esophagus involvement (1390±134.4 vs 940.7±127.0 ng/ml, P=0.0269; TGF-β, 1.9±0.3 vs 0.9±0.2 ng/ml, P=0.0426). The area under receiver operating characteristics (ROC) curve of LTBP-4 was 0.86. Immunohistochemistry measurement also demonstrated a higher LTBP-4 expression in sclerotic skin tissue of LSc and SSc compared with normal controls. More positive fibroblasts were also found in BLM-induced scleroderma mouse model than the saline-treated group. In in vitro studies, knockdown of LTBP-4 in SSc skin fibroblasts prominently reduced downstream COL1A1, COL1A2, and COL3A1 mRNA level by 84%, 82%, and 43%, respectively, and other fibrosis-related genes' expression were also decreased. Furthermore, extracellular TGF-β level and the SMAD2/3 phosphorylation were inhibited through LTBP-4 knockdown treatment, suggesting that the knockdown of LTBP-4 reduced the collagen expression through TGF-β/SMAD signaling pathway. Taken together, these data suggest that LTBP-4 affects fibrotic process in scleroderma, and the high expression of LTBP-4 in SSc plasma may serve as a clinical biomarker in diagnosing this disease. In addition, this study also lays the theoretical foundation for targeting LTBP-4 as treatment of scleroderma.
Collapse
Affiliation(s)
- Jiaying Lu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingmei Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.,State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Lei Wang
- Division of Rheumatology, Shanghai TCM-integrated Hospital, Shanghai, China
| | - Wenzhen Tu
- Division of Rheumatology, Shanghai TCM-integrated Hospital, Shanghai, China
| | - Haiyan Chu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weifeng Ding
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiangguang Shi
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weilin Pu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaodong Zhou
- University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Nabissi M, Morelli MB, Amantini C, Liberati S, Santoni M, Ricci-Vitiani L, Pallini R, Santoni G. Cannabidiol stimulates Aml-1a-dependent glial differentiation and inhibits glioma stem-like cells proliferation by inducing autophagy in a TRPV2-dependent manner. Int J Cancer 2015; 137:1855-69. [PMID: 25903924 DOI: 10.1002/ijc.29573] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/15/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Massimo Nabissi
- Section of Experimental Medicine, School of Pharmacy; University of Camerino; Camerino Italy
| | - Maria Beatrice Morelli
- Section of Experimental Medicine, School of Pharmacy; University of Camerino; Camerino Italy
- Department of Molecular Medicine; Sapienza University; Rome Italy
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine; University of Camerino; Camerino Italy
| | - Sonia Liberati
- Department of Molecular Medicine; Sapienza University; Rome Italy
| | - Matteo Santoni
- Clinica Di Oncologia Medica; AOU Ospedali Riuniti-Università Politecnica Delle Marche; Ancona Italy
| | - Lucia Ricci-Vitiani
- Department of Hematology, Oncology and Molecular Medicine; Istituto Superiore Di Sanità; Rome Italy
| | - Roberto Pallini
- Department of Neurosurgery; Università Cattolica Del Sacro Cuore; Rome Italy
| | - Giorgio Santoni
- Section of Experimental Medicine, School of Pharmacy; University of Camerino; Camerino Italy
| |
Collapse
|
14
|
Ohta N, Ishiguro S, Kawabata A, Uppalapati D, Pyle M, Troyer D, De S, Zhang Y, Becker KG, Tamura M. Human umbilical cord matrix mesenchymal stem cells suppress the growth of breast cancer by expression of tumor suppressor genes. PLoS One 2015; 10:e0123756. [PMID: 25942583 PMCID: PMC4420498 DOI: 10.1371/journal.pone.0123756] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/24/2015] [Indexed: 12/25/2022] Open
Abstract
Human and rat umbilical cord matrix mesenchymal stem cells (UCMSC) possess the ability to control the growth of breast carcinoma cells. Comparative analyses of two types of UCMSC suggest that rat UCMSC-dependent growth regulation is significantly stronger than that of human UCMSC. Their different tumoricidal abilities were clarified by analyzing gene expression profiles in the two types of UCMSC. Microarray analysis revealed differential gene expression between untreated naïve UCMSC and those co-cultured with species-matched breast carcinoma cells. The analyses screened 17 differentially expressed genes that are commonly detected in both human and rat UCMSC. The comparison between the two sets of gene expression profiles identified two tumor suppressor genes, adipose-differentiation related protein (ADRP) and follistatin (FST), that were specifically up-regulated in rat UCMSC, but down-regulated in human UCMSC when they were co-cultured with the corresponding species' breast carcinoma cells. Over-expression of FST, but not ADRP, in human UCMSC enhanced their ability to suppress the growth of MDA-231 cells. The growth of MDA-231 cells was also significantly lower when they were cultured in medium conditioned with FST, but not ADRP over-expressing human UCMSC. In the breast carcinoma lung metastasis model generated with MDA-231 cells, systemic treatment with FST-over-expressing human UCMSC significantly attenuated the tumor burden. These results suggest that FST may play an important role in exhibiting stronger tumoricidal ability in rat UCMSC than human UCMSC and also implies that human UCMSC can be transformed into stronger tumoricidal cells by enhancing tumor suppressor gene expression.
Collapse
Affiliation(s)
- Naomi Ohta
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Susumu Ishiguro
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Atsushi Kawabata
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Deepthi Uppalapati
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Marla Pyle
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Deryl Troyer
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Supriyo De
- Gene Expression and Genomics Unit, NIH Biomedical Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224, United States of America
| | - Yongqing Zhang
- Gene Expression and Genomics Unit, NIH Biomedical Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224, United States of America
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, NIH Biomedical Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224, United States of America
| | - Masaaki Tamura
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
- * E-mail:
| |
Collapse
|
15
|
Chistiakov DA, Chekhonin VP. Extracellular vesicles shed by glioma cells: pathogenic role and clinical value. Tumour Biol 2014; 35:8425-38. [DOI: 10.1007/s13277-014-2262-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/18/2014] [Indexed: 02/03/2023] Open
|
16
|
Bultmann I, Conradi A, Kretschmer C, Sterner-Kock A. Latent transforming growth factor β-binding protein 4 is downregulated in esophageal cancer via promoter methylation. PLoS One 2013; 8:e65614. [PMID: 23741501 PMCID: PMC3669142 DOI: 10.1371/journal.pone.0065614] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/26/2013] [Indexed: 02/07/2023] Open
Abstract
Latent transforming growth factor β-binding protein 4 (LTBP4) is an extracellular matrix molecule that is a member of important connective tissue networks and is needed for the correct folding and the secretion of TGF-β1. LTBP4 is downregulated in carcinomas of various tissues. Here we show that LTBP4 is also downregulated in adenocarcinomas and squamous cell carcinomas of the esophagus in vitro and in vivo. Re-expression of LTBP4 in esophageal cancer cell lines reduced cell migration ability, whereas cell viability and cell proliferation remained unchanged. Hypermethylation of the promoter regions of the two main human LTBP4 transcriptional forms, LTBP4L and LTBP4S, was found to be involved in LTBP4 silencing. Detailed investigations of the methylation patterns of the promoter regions of LTBP4L and LTBP4S identified GATA1, SP1, E2F4 and SMAD3 as potential transcription factors involved in LTBP4 expression. In in vitro transcription factor activity studies we discovered E2F4 as novel powerful regulator for LTBP4S expression.
Collapse
Affiliation(s)
- Insa Bultmann
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | | | | | | |
Collapse
|
17
|
Lu X, Wang Y, Luo H, Qiu W, Han H, Chen X, Yang L. β-elemene inhibits the proliferation of T24 bladder carcinoma cells through upregulation of the expression of Smad4. Mol Med Rep 2012; 7:513-8. [PMID: 23228961 DOI: 10.3892/mmr.2012.1206] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 11/05/2012] [Indexed: 11/06/2022] Open
Abstract
β-elemene, a non-cytotoxic antitumor reagent, inhibits the growth, proliferation and DNA synthesis of multiple types of malignant cells, resulting in the apoptosis or suppressed vascularization of tumors. β-elemene is also able to enhance the immunogenicity of the tumor cells and ameliorate systematic cellular immunity in the tumor‑bearing body. Moreover, β-elemene has the advantages of high efficiency, safety and low possibility of drug tolerance over other antitumor agents used in antitumor treatment. Therefore, β-elemene has great potential in clinical applications. However, the mechanism of β-elemene antitumor activity is largely unknown. The aim of this study was to investigate whether β-elemene is able to repress the proliferation of T24 bladder carcinoma cells through regulation of the expression of the tumor suppressor gene, Smad4. Results of a methylthiazolyl tetrazolium (MTT) assay indicated that the proliferation of T24 cells was repressed by β-elemene in a time- and concentration‑dependent manner. The lowest concentration of β-elemene to inhibit cell survival by >50% was determined using IC50 software. Microscopic observation also demonstrated the potential of β-elemene to induce the apoptosis of cancer cells. Western blot and RT-PCR analyses revealed that the expression of the Smad4 protein and mRNA was upregulated by treatment with β-elemene. Our results revealed that β-elemene was able to upregulate the expression of Smad4 in tumor cells to inhibit the proliferation of these cells.
Collapse
Affiliation(s)
- Xinsheng Lu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | | | | | | | | | | | | |
Collapse
|