1
|
Balogh G, Jorge N, Dupain C, Kamal M, Servant N, Le Tourneau C, Stadler PF, Bernhart SH. TREMSUCS-TCGA - an integrated workflow for the identification of biomarkers for treatment success. J Integr Bioinform 2024; 21:jib-2024-0031. [PMID: 39654143 PMCID: PMC11698617 DOI: 10.1515/jib-2024-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/18/2024] [Indexed: 01/06/2025] Open
Abstract
Many publicly available databases provide disease related data, that makes it possible to link genomic data to medical and meta-data. The cancer genome atlas (TCGA), for example, compiles tens of thousand of datasets covering a wide array of cancer types. Here we introduce an interactive and highly automatized TCGA-based workflow that links and analyses epigenomic and transcriptomic data with treatment and survival data in order to identify possible biomarkers that indicate treatment success. TREMSUCS-TCGA is flexible with respect to type of cancer and treatment and provides standard methods for differential expression analysis or DMR detection. Furthermore, it makes it possible to examine several cancer types together in a pan-cancer type approach. Parallelisation and reproducibility of all steps is ensured with the workflowmanagement system Snakemake. TREMSUCS-TCGA produces a comprehensive single report file which holds all relevant results in descriptive and tabular form that can be explored in an interactive manner. As a showcase application we describe a comprehensive analysis of the available data for the combination of patients with squamous cell carcinomas of head and neck, cervix and lung treated with cisplatin, carboplatin and the combination of carboplatin and paclitaxel. The best ranked biomarker candidates are discussed in the light of the existing literature, indicating plausible causal relationships to the relevant cancer entities.
Collapse
Affiliation(s)
- Gabor Balogh
- Interdisciplinary Center of Bioinformatics, Leipzig University, Härtelstraße 16-18, D-04107Leipzig, Germany
- Bioinformatics Group, Institute of Computer Science, Leipzig University, Härtelstraße 16-18, D-04107Leipzig, Germany
| | - Natasha Jorge
- Interdisciplinary Center of Bioinformatics, Leipzig University, Härtelstraße 16-18, D-04107Leipzig, Germany
- Bioinformatics Group, Institute of Computer Science, Leipzig University, Härtelstraße 16-18, D-04107Leipzig, Germany
| | - Célia Dupain
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | | | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- Inserm U900 Research Unit, Saint Cloud, France
- Université Paris-Saclay, 91190Gif-sur-Yvette, France
| | - Peter F. Stadler
- Interdisciplinary Center of Bioinformatics, Leipzig University, Härtelstraße 16-18, D-04107Leipzig, Germany
- Bioinformatics Group, Institute of Computer Science, Leipzig University, Härtelstraße 16-18, D-04107Leipzig, Germany
- Max-Planck-Institute for Mathematics in Sciences, Inselstraße 22, D-04109Leipzig, Germany
- Leipzig Research Center for Civilization Diseases (LIFE), University Leipzig, Härtelstrasse 16-18, D-04107Leipzig, Germany
- Department of Theoretical Chemistry of the University of Vienna, Währingerstrasse 17, A-1090 Vienna, Austria
- Center for RNA in Technology and Health (RTH), Univ. Copenhagen, Grønnegårdsvej 3, Frederiksberg C, Denmark
- Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá CO-111321, Colombia
- Santa Fe Institute, 1399 Hyde Park Road, Santa Fe NM 87501, USA
| | - Stephan H. Bernhart
- Interdisciplinary Center of Bioinformatics, Leipzig University, Härtelstraße 16-18, D-04107Leipzig, Germany
- Bioinformatics Group, Institute of Computer Science, Leipzig University, Härtelstraße 16-18, D-04107Leipzig, Germany
| |
Collapse
|
2
|
Lin J, Wang D, Zhou J, Bai J, Sun S, Jia X, Liang X, Fu S, Yu J. MIEN1 on the 17q12 amplicon facilitates the malignant behaviors of gastric cancer via activating IL-6/JAK2/STAT3 pathway. Int J Biochem Cell Biol 2024; 176:106666. [PMID: 39343060 DOI: 10.1016/j.biocel.2024.106666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/03/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
Oncogene amplification is a significant factor contributing to poor prognosis and limited treatment in patients with advanced gastric cancer. Therefore, identifying amplified oncogenes and elucidating their oncogenic mechanisms will provide reliable therapeutic targets for the clinical treatment of gastric cancer. In this study, we identify a high amplification of 17q12, which includes five oncogenes that are co-amplified and co-overexpressed with ERBB2 using array comparative genomic hybridization, with migration and invasion enhancer 1 (MIEN1) being particularly highlighted for its clinical significance, function, and role in gastric cancer progression. By detecting MIEN1 copy number and expression level across eight gastric cancer cell lines and in tissue microarrays from 543 primary gastric cancer tissues, we found that MIEN1 amplification and overexpression correlated with sex and Lauren's intestinal type classification of gastric cancer. Besides that, elevated MIEN1 expression was associated with poorer patient survival. In vitro experiments have shown that MIEN1 overexpression enhanced cell proliferation, invasion, and migration, whereas MIEN1 knockdown reversed these malignant phenotypes in vitro. Furthermore, MIEN1 knockdown inhibited tumorigenesis and metastasis of gastric cancer cells in nude mice. Mechanistically, MIEN1 activates the IL-6/JAK2/STAT3 signaling pathway, which drives the proliferation, invasion, and migration of gastric cancer cells. This study demonstrates that MIEN1 contributes to the malignant behavior of gastric cancer through the IL-6/JAK2/STAT3 pathway, suggesting that MIEN1 could serve as a valuable therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Jing Lin
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Dong Wang
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Jiahui Zhou
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Jing Bai
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Shouzhen Sun
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Xueyuan Jia
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Xiao Liang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Songbin Fu
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Jingcui Yu
- Scientific Research Centre, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China.
| |
Collapse
|
3
|
Fang W, Chen S, Wan D, Peng Y, Yang X. Identification and Validation of an Invasion-Related Disease-Free Survival Prognostic Model for Tongue Squamous Cell Carcinoma. Oncology 2024; 103:237-252. [PMID: 39307124 DOI: 10.1159/000540977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 08/14/2024] [Indexed: 03/08/2025]
Abstract
INTRODUCTION Tongue squamous cell carcinoma (TSCC) is a common malignant tumour type with aggressive invasion and a poor prognosis. To date, invasion-related gene expression signatures for the prognostic stratification of TSCC patients are unavailable in clinical practice. This study aimed to assess the impact of invasion-related genes on the prognosis of TSCC patients. METHODS We obtained mRNA profiles and clinical data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases (TCGA-TSCC and GSE41116, respectively). The TSCC samples from the TCGA-TSCC cohort were randomly divided into TCGA training and TCGA test datasets at a 7:3 ratio. Next, a disease-free survival (DFS) prognostic risk model was established on the basis of univariate and stepwise multivariate Cox regression analyses of the TCGA training cohort. Moreover, prognostic genes were screened. The model was subsequently evaluated and validated using the TCGA test and GSE41116 datasets. In addition, the prognostic genes were validated in the human TSCC cell line UM1 and the human oral keratinocyte (HOK) cell line using quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RESULTS A total of 70 candidate genes related to invasion were identified in the TCGA-TSCC cohort. DFS data were subsequently constructed, and 6 prognostic genes, HMGN2, MYL12B, ACTB, PPP1CA, PSMB9, and IFITM3, were identified. The TSCC samples were divided into high- and low-risk groups in the TCGA training, TCGA test, and GSE41116 cohorts, respectively. In particular, patients with TSCC in the low-risk group had longer DFS than those in the high-risk group. Furthermore, qRT-PCR analysis confirmed that the expression levels of the 6 prognostic genes were significantly greater in the TSCC cell line UM1 than in the HOK cell line. CONCLUSION This study identified new invasion-related target genes related to poor prognosis in TSCC patients, providing new insights into the underlying mechanisms of TSCC invasion.
Collapse
Affiliation(s)
- Wei Fang
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Shan Chen
- Department of Stomatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Di Wan
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yanhui Peng
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoqin Yang
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Heidarzadehpilehrood R, Pirhoushiaran M. Biomarker potential of competing endogenous RNA networks in Polycystic Ovary Syndrome (PCOS). Noncoding RNA Res 2024; 9:624-640. [PMID: 38571815 PMCID: PMC10988127 DOI: 10.1016/j.ncrna.2024.01.002] [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: 10/22/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 04/05/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common condition affecting women of reproductive age globally. PCOS continues to be the largest contributing factor to female infertility despite significant progress in our knowledge of the molecular underpinnings and treatment of the condition. The fact that PCOS is a very diverse condition makes it one of the key reasons why we haven't been able to overcome it. Non-coding RNAs (ncRNAs) are implicated in the development of PCOS, according to growing evidence. However, it is unclear how the complex regulatory relationships between the many ncRNA types contribute to the growth of this malignancy. Competing endogenous RNA (ceRNA), a recently identified mechanism in the RNA world, suggests regulatory interactions between various RNAs, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), transcribed pseudogenes, and circular RNAs (circRNAs). Recent studies on PCOS have shown that dysregulation of multiple ceRNA networks (ceRNETs) between these ncRNAs plays crucial roles in developing the defining characteristics of PCOS development. And it is believed that such a finding may open a new door for a deeper comprehension of PCOS's unexplored facets. In addition, it may be able to provide fresh biomarkers and effective therapy targets for PCOS. This review will go over the body of information that exists about the primary roles of ceRNETs before highlighting the developing involvement of several newly found ceRNETs in a number of PCOS characteristics.
Collapse
Affiliation(s)
- Roozbeh Heidarzadehpilehrood
- Department of Obstetrics & Gynaecology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Maryam Pirhoushiaran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, 1417613151, Iran
| |
Collapse
|
5
|
Zeng C, Guo J, Wu J, Che T, Huang X, Liu H, Lin Z. HRD1 Promotes Non-small Cell Lung Carcinoma Metastasis by Blocking Autophagy-mediated MIEN1 Degradation. J Biol Chem 2023; 299:104723. [PMID: 37075843 DOI: 10.1016/j.jbc.2023.104723] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 04/21/2023] Open
Abstract
Dysregulation of autophagy has been implicated in the development of many diseases, including cancer. Here, we revealed a novel function of the E3 ubiquitin ligase HRD1 in non-small cell lung carcinoma (NSCLC) metastasis by regulating autophagy. Mechanistically, HRD1 inhibits autophagy by promoting ATG3 ubiquitination and degradation. Additionally, a pro-migratory and invasive factor, MIEN1 (migration and invasion enhancer 1), was found to be autophagically degraded upon HRD1 deficiency. Importantly, both HRD1 and MIEN1 expression are upregulated and positively correlated in lung tumors. Based on these results, we proposed a novel mechanism of HRD1 function that the degradation of ATG3 protein by HRD1 leads to autophagy inhibition and MIEN1 release, thus promoting NSCLC metastasis. Therefore, our findings provided new insights into the role of HRD1 in NSCLC metastasis and new therapeutic targets for lung cancer treatment.
Collapse
Affiliation(s)
- Cheng Zeng
- School of Life Sciences, Chongqing University, Chongqing 401331, P.R. China
| | - Jing Guo
- Affiliated Three Gorges Central Hospital of Chongqing University, Chongqing, P. R. China
| | - Jiajia Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, P.R. China
| | - Tiantian Che
- School of Life Sciences, Chongqing University, Chongqing 401331, P.R. China
| | - Xiaoping Huang
- Affiliated Three Gorges Central Hospital of Chongqing University, Chongqing, P. R. China
| | - Huawen Liu
- Affiliated Three Gorges Central Hospital of Chongqing University, Chongqing, P. R. China.
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, P.R. China.
| |
Collapse
|
6
|
Associations of selenoprotein expression and gene methylation with the outcome of clear cell renal carcinoma. Arch Biochem Biophys 2023; 733:109470. [PMID: 36442530 DOI: 10.1016/j.abb.2022.109470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
Selenoproteins are a ubiquitous class of proteins defined by having a selenocysteine amino acid residue. While many of the selenoproteins have been well characterized with important roles in oxidation-reduction reactions and hormone synthesis among others, there exist some whose biological roles are not as well understood as denoted by the "SELENO" root. In this study, we explored associations between the reported RNA levels of "SELENO" proteins and clear cell renal cell carcinoma (ccRCC), the most common subtype of renal carcinoma in the US. Utilizing The Cancer Genome Atlas (TCGA) alongside other in silico tools, we discovered higher mRNA expression of Selenoprotein I, T, and P was associated with better overall survival outcomes and differential expression of other selenoproteins based on tumor stage. Additionally, we uncovered relative hypomethylation among selenoproteins in primary ccRCC tumor samples compared to normal tissue. Network and enrichment analysis showed numerous genes through which selenoproteins may modulate cancer progression and outcomes such as DERL1, PNPLA2/3, MIEN1, and FOXO1 which have been well-described in other cancers. In light of our findings highlighting an association of selenoprotein methylation and expression patterns with ccRCC outcome, further wet lab research is warranted.
Collapse
|
7
|
Terkelsen T, Pernemalm M, Gromov P, Børresen-Dale AL, Krogh A, Haakensen VD, Lethiö J, Papaleo E, Gromova I. High-throughput proteomics of breast cancer interstitial fluid: identification of tumor subtype-specific serologically relevant biomarkers. Mol Oncol 2021; 15:429-461. [PMID: 33176066 PMCID: PMC7858121 DOI: 10.1002/1878-0261.12850] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/13/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022] Open
Abstract
Despite significant advancements in breast cancer (BC) research, clinicians lack robust serological protein markers for accurate diagnostics and tumor stratification. Tumor interstitial fluid (TIF) accumulates aberrantly externalized proteins within the local tumor space, which can potentially gain access to the circulatory system. As such, TIF may represent a valuable starting point for identifying relevant tumor-specific serological biomarkers. The aim of the study was to perform comprehensive proteomic profiling of TIF to identify proteins associated with BC tumor status and subtype. A liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis of 35 TIFs of three main subtypes: luminal (19), Her2 (4), and triple-negative (TNBC) (12) resulted in the identification of > 8800 proteins. Unsupervised hierarchical clustering segregated the TIF proteome into two major clusters, luminal and TNBC/Her2 subgroups. High-grade tumors enriched with tumor infiltrating lymphocytes (TILs) were also stratified from low-grade tumors. A consensus analysis approach, including differential abundance analysis, selection operator regression, and random forest returned a minimal set of 24 proteins associated with BC subtypes, receptor status, and TIL scoring. Among them, a panel of 10 proteins, AGR3, BCAM, CELSR1, MIEN1, NAT1, PIP4K2B, SEC23B, THTPA, TMEM51, and ULBP2, was found to stratify the tumor subtype-specific TIFs. In particular, upregulation of BCAM and CELSR1 differentiates luminal subtypes, while upregulation of MIEN1 differentiates Her2 subtypes. Immunohistochemistry analysis showed a direct correlation between protein abundance in TIFs and intratumor expression levels for all 10 proteins. Sensitivity and specificity were estimated for this protein panel by using an independent, comprehensive breast tumor proteome dataset. The results of this analysis strongly support our data, with eight of the proteins potentially representing biomarkers for stratification of BC subtypes. Five of the most representative proteomics databases currently available were also used to estimate the potential for these selected proteins to serve as putative serological markers.
Collapse
Affiliation(s)
- Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Maria Pernemalm
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Pavel Gromov
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anna-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Anders Krogh
- Department of Computer Science, University of Copenhagen, Denmark.,Department of Biology, University of Copenhagen, Denmark
| | - Vilde D Haakensen
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Janne Lethiö
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark.,Translational Disease System Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Denmark
| | - Irina Gromova
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| |
Collapse
|
8
|
Liang F, Zhang H, Qiu Y, Xu Q, Jian K, Jiang L, Wang F, Lu X. MiR-124-5p Inhibits the Progression of Gastric Cancer by Targeting MIEN1. Technol Cancer Res Treat 2020; 19:1533033820979199. [PMID: 33349155 PMCID: PMC7758558 DOI: 10.1177/1533033820979199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective: To observe the effect of miR-124-5p on progression of gastric cancer (GC) and explore the targeting mechanism. Methods: After collecting the specimens, we used real-time fluorescence quantitative PCR to detect the miR-124-5p level of GC tissue and corresponding adjacent tissue. Then MTT test and scratch wound-healing assay were hired to evaluate the influence of miR-124-5p in GC cell (SGC-803 and SGC7901) migration and proliferation ability. The binding of miR-124-5p to migration and invasion enhancer 1 (MIEN1) was detected through dual luciferase reporter gene experiment and western blot was utilized to assay the protein level of MIEN1. Results: Compared with adjacent tissues, miR-124-5p level in GC tissues was lower significantly. MiR-124-5p mimic inhibited the metastasis and proliferation ability of SGC7901 cells and miR-124-5p inhibitor promoted the migration and proliferation ability of SGC803 cells. In addition, miR-124-5p targeted MIEN1 and negatively modulated the MIEN1 expression in SGC-803 and SGC7901 cells. Silencing MIEN1 negatively regulated the metastasis and proliferation ability of SGC7901 cells. Conclusion: MiR-124-5p inhibited the GC cell proliferation and metastasis phenotypes through MIEN1, which probably becomes a novel molecular target for clinical GC treatment.
Collapse
Affiliation(s)
- Feng Liang
- Department of General Surgery, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - HongYan Zhang
- Department of Oncology, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - YuXuan Qiu
- Department of General Surgery, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - QianRu Xu
- Department of General Surgery, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - KaiYu Jian
- Department of General Surgery, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Lin Jiang
- Department of General Surgery, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Fei Wang
- Department of Oncology, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Xin Lu
- Department of General Surgery, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
9
|
Yu X, Xiao W, Song H, Jin Y, Xu J, Liu X. CircRNA_100876 sponges miR-136 to promote proliferation and metastasis of gastric cancer by upregulating MIEN1 expression. Gene 2020; 748:144678. [PMID: 32305633 DOI: 10.1016/j.gene.2020.144678] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/30/2020] [Accepted: 04/14/2020] [Indexed: 01/17/2023]
Abstract
Previous reports have revealed that circRNA_100876 was extremely important in the progression of triple-negative breast cancer. Nevertheless, the mechanism towards the role of circRNA_100876 in Gastric cancer (GC) remains unknown. Here, we determined circRNA_100876 expression by quantitative real-time PCR (qRT-PCR) in twenty pairs of GC tissues and adjacent tissues. Our data indicated that the expression of circRNA_100876 was raised in GC tissues. In vitro, functional experiments confirmed that cell proliferation, invasion along with migration was promoted by circRNA_100876 in GC tissues. Simultaneously, relative luciferase assay uncovered that circRNA_100876 functioned as a sponge for miR-136, followed by retarding miR-136-induced inhibited effects on the corresponding target, MIEN1. Moreover, we revealed that the expression of MIEN1 was up-regulated and correlated to much worse prognosis of GC. Collectively, our data identified that the promotion of GC growth and metastasis induced by circRNA_100876 interacted with miR-136 and MIEN1, indicating an emerging announcement for uncovering the potential mechanism of GC progression.
Collapse
Affiliation(s)
- Xiaoyun Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Wenjing Xiao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Hao Song
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yonglong Jin
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Jinpeng Xu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiguang Liu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
10
|
Migration and Invasion Enhancer 1 Is an NF-ĸB-Inducing Gene Enhancing the Cell Proliferation and Invasion Ability of Human Prostate Carcinoma Cells In Vitro and In Vivo. Cancers (Basel) 2019; 11:cancers11101486. [PMID: 31581708 PMCID: PMC6826896 DOI: 10.3390/cancers11101486] [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: 09/06/2019] [Revised: 09/24/2019] [Accepted: 09/30/2019] [Indexed: 12/22/2022] Open
Abstract
Migration and invasion enhancer 1 (MIEN1) is a membrane-anchored protein and exists in various cancerous tissues. However, the roles of MIEN1 in prostate cancer have not yet been clearly addressed. We determined the expression, biological functions, and regulatory mechanisms of MIEN1 in the prostate. The results of immunohistochemical analysis indicated that MIEN1 was expressed specifically in epithelial cells and significantly higher in adenocarcinoma as compared to in normal tissues. MIEN1 enhanced in vitro cell proliferation, invasion, and in vivo tumorigenesis. Meanwhile, MIEN1 attenuated cisplatin-induced apoptosis in PC-3 cells. Overexpression of NF-ĸB-inducing kinase (NIK) enhanced MIEN1 expression, while overexpression of NF-ĸB inhibitor α (IĸBα) blocked MIEN1 expression in PC-3 cells. In prostate carcinoma cells, MIEN1 provoked Akt phosphorylation; moreover, MIEN1 downregulated N-myc downstream regulated 1 (NDRG1) but upregulated interleukin-6 (IL-6) gene expression. MK2206, an Akt inhibitor, impeded the modulation of MIEN1 on NDRG1 and IL-6 expressions. Our studies suggest that MIEN1 is an NF-ĸB downstream oncogene in the human prostate. Accordingly, the modulation of Akt signaling in the gene expressions of NDRG1 and IL-6 may account for the functions of MIEN1 in cell proliferation, invasion, and tumorigenesis in prostate carcinoma cells.
Collapse
|
11
|
Kushwaha PP, Gupta S, Singh AK, Kumar S. Emerging Role of Migration and Invasion Enhancer 1 (MIEN1) in Cancer Progression and Metastasis. Front Oncol 2019; 9:868. [PMID: 31552186 PMCID: PMC6738349 DOI: 10.3389/fonc.2019.00868] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/21/2019] [Indexed: 02/05/2023] Open
Abstract
Tumor metastasis is a sequential event accounting for numerous cancer-related fatalities worldwide. The process of metastasis serially involves invasion, intravasation, extravasation, and tumor growth at the secondary site. Migration and invasion enhancer 1 (MIEN1) is a membrane associated protein overexpressed in various human cancers. Biological activity of MIEN1 is driven by geranylgeranyltransferase-I mediated prenylation at CAAX motif and methylation of the prenylated protein that anchors MIEN1 into the cellular membrane. Post-translationally modified MIEN1 interacts with Syk kinase and Annexin A2 protein; polymerizes G-actin and stabilizes F-actin filament; induces focal adhesion kinase phosphorylation and decrease cofilin phosphorylation implicated in both invasion and metastasis of different cancer types. In the present review, we discuss the structure, function, and involvement of MIEN1 in cancer progression. We also highlight the future prospects of MIEN1 as an emerging molecule and novel target in cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Prem Prakash Kushwaha
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, OH, United States
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
- Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH, United States
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Atul Kumar Singh
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Shashank Kumar
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
12
|
Feghali KAA, Ghanem AI, Burmeister C, Chang SS, Ghanem T, Keller C, Siddiqui F. Impact of smoking on pathological features in oral cavity squamous cell carcinoma. J Cancer Res Ther 2019; 15:582-588. [PMID: 31169224 PMCID: PMC7470189 DOI: 10.4103/jcrt.jcrt_641_16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVES We sought to determine whether smokers with oral cavity squamous cell carcinoma (OCSCC) have tumors with more adverse pathological features than in nonsmokers and whether or not these are predictive of outcomes. MATERIALS AND METHODS We retrospectively identified 163 patients with American Joint Committee on Cancer stages I-IVa OCSCC diagnosed between 2005 and 2015 and treated with curative intent. A pathological risk score (PRS) was calculated using the National Comprehensive Cancer Network adverse risk factors: positive margin, extracapsular extension of lymph node metastases, pT3 or pT4 primary, N2 or N3 nodal disease, perineural invasion, and lymphovascular space invasion. Multivariable models were constructed to determine the independent predictors of overall survival (OS), recurrence-free survival (RFS), and PRS. RESULTS A total of 108 (66.26%) were smokers and 55 nonsmokers. Three-year actuarial OS and RFS were 62% and 68% in smokers and 81% and 69% in nonsmokers, respectively (P = 0.06 and P = 0.63). Smokers were more likely to have advanced disease stage and tumors with aggressive pathological features than nonsmokers. Smokers had significantly worse PRS (mean ± standard deviation; 2.38 ± 2.19, median; 2.00) than nonsmokers (0.89 ± 1.21, 0.00) (P < 0.001). Older age, higher PRS, and smoking status were independent predictors of OS. Smoking or PRS did not predict for worse RFS. On multivariate analysis, independent predictors of PRS were smoking status and grade (P < 0.001). CONCLUSION In patients with OCSCC, smokers have more aggressive disease as evidenced by more adverse pathological features than nonsmokers. Moreover, smoking is an independent predictor of OS but not RFS. The PRS is a significant predictor of OS and needs validation in the future studies.
Collapse
Affiliation(s)
| | - Ahmed I. Ghanem
- Department of Radiation Oncology, Henry Ford Hospital, Detroit, MI, USA
| | | | - Steven S. Chang
- Department of Otolaryngology - Head and Neck Surgery, Henry Ford Hospital, Detroit, MI, USA
| | - Tamer Ghanem
- Department of Otolaryngology - Head and Neck Surgery, Henry Ford Hospital, Detroit, MI, USA
| | | | - Farzan Siddiqui
- Department of Radiation Oncology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
13
|
Kurtz P, Jones AE, Tiwari B, Link N, Wylie A, Tracy C, Krämer H, Abrams JM. Drosophila p53 directs nonapoptotic programs in postmitotic tissue. Mol Biol Cell 2019; 30:1339-1351. [PMID: 30892991 PMCID: PMC6724604 DOI: 10.1091/mbc.e18-12-0791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 12/26/2022] Open
Abstract
TP53 is the most frequently mutated gene in human cancers, and despite intensive research efforts, genome-scale studies of p53 function in whole animal models are rare. The need for such in vivo studies is underscored by recent challenges to established paradigms, indicating that unappreciated p53 functions contribute to cancer prevention. Here we leveraged the Drosophila system to interrogate p53 function in a postmitotic context. In the developing embryo, p53 robustly activates important apoptotic genes in response to radiation-induced DNA damage. We recently showed that a p53 enhancer (p53RErpr) near the cell death gene reaper forms chromatin contacts and enables p53 target activation across long genomic distances. Interestingly, we found that this canonical p53 apoptotic program fails to activate in adult heads. Moreover, this failure to exhibit apoptotic responses was not associated with altered chromatin contacts. Instead, we determined that p53 does not occupy the p53RErpr enhancer in this postmitotic tissue as it does in embryos. Through comparative RNA-seq and chromatin immunoprecipitation-seq studies of developing and postmitotic tissues, we further determined that p53 regulates distinct transcriptional programs in adult heads, including DNA repair, metabolism, and proteolysis genes. Strikingly, in the postmitotic context, p53-binding landscapes were poorly correlated with nearby transcriptional effects, raising the possibility that p53 enhancers could be generally acting through long distances.
Collapse
Affiliation(s)
- Paula Kurtz
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Amanda E. Jones
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Bhavana Tiwari
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nichole Link
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Houston, TX 77030
| | - Annika Wylie
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Charles Tracy
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Helmut Krämer
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John M. Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
14
|
Chu Y, Hu X, Wang G, Wang Z, Wang Y. Downregulation of miR-136 promotes the progression of osteosarcoma and is associated with the prognosis of patients with osteosarcoma. Oncol Lett 2019; 17:5210-5218. [PMID: 31186737 DOI: 10.3892/ol.2019.10203] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/02/2019] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone tumor in children and young adults, and is an aggressive tumor with poor prognosis. MicroRNAs (miRNAs) are aberrantly expressed in various types of cancer, and contribute to cancer tumorigenesis and progression. In the present study, the potential prognostic value and biological function of miRNA-136 (miR-136) in OS was investigated. Reverse transcription-quantitative polymerase chain reaction analysis was used to evaluate the expression of miR-136 in OS tissues and cell lines. Kaplan-Meier survival analysis and Cox regression analysis were conducted to investigate the prognostic significance of miR-136. Various in vitro cell based assays were used to evaluate the effects of miR-136 on the biological behavior of OS cells. A luciferase assay was performed to determine the key miR-136 targets associated with OS. The expression of miR-136 was significantly downregulated in osteosarcoma tissues and cells compared with the normal controls (all P<0.05). Decreased miR-136 expression was significantly associated with Enneking staging (P=0.030) and distant metastasis (P=0.016). Decreased miR-136 expression in patients was associated with shorter overall survival compared with patients with increased expression levels (log-rank test; P<0.05). The expression of miR-136 was indicated as an independent prognostic factor for the patients (hazard ratio=0.496; 95% confidence interval=0.250-0.987; P=0.046). MTT, transwell and Matrigel assays demonstrated that upregulation of miR-136 decreased proliferation, migration and invasion of OS cells. Bioinformatics and luciferase assays demonstrated that migration and invasion enhancer 1 (MIEN1) is a direct target of miR-136. Together, the results suggested that miR-136 functions as a tumor suppressor gene to regulate proliferation, migration and invasion of OS cells. MIEN1 was a potential target of miR-136. Additionally, miR-136 may serve as a prognostic biomarker for OS.
Collapse
Affiliation(s)
- Yanchen Chu
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Xiaoli Hu
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Linyi City, Linyi, Shandong 276000, P.R. China
| | - Guangfeng Wang
- Department of Medical Administration, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Zhijie Wang
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Yanjin Wang
- Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
15
|
Van Treuren T, Vishwanatha JK. CRISPR deletion of MIEN1 in breast cancer cells. PLoS One 2018; 13:e0204976. [PMID: 30286132 PMCID: PMC6171864 DOI: 10.1371/journal.pone.0204976] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 09/18/2018] [Indexed: 12/26/2022] Open
Abstract
Migration and Invasion Enhancer (MIEN1) is an oncogene which is involved in facilitating motility of cancer cells through actin dynamics and gene expression. Increased MIEN1 expression in many types of tumors leads to disease progression and metastatic propensity. It is unclear precisely how MIEN1 is involved in this process and more studies are required to tease out the mechanisms. Here we show that Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR) genome editing effectively produced specific genomic deletions in the MIEN1 gene which led to the abrogation of its expression in breast cancer cells. The single guide RNAs (sgRNAs) mediated targeting of MIEN1 was specific and none of the clones screened for off-target cleavage revealed any insertions or deletions (indels). Additionally, disruption of the MIEN1 gene did not alter the cell morphology, growth, proliferation or survival. Knocking out MIEN1 in these breast cancer cells will allow future studies to determine the exact role MIEN1 plays in breast tumor metastasis, which might lead to production of novel therapeutics to treat this and other cancers.
Collapse
Affiliation(s)
- Timothy Van Treuren
- Department of Microbiology, Immunology and Genetics, Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- * E-mail:
| | - Jamboor K. Vishwanatha
- Department of Microbiology, Immunology and Genetics, Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| |
Collapse
|
16
|
Bierbaumer L, Schwarze UY, Gruber R, Neuhaus W. Cell culture models of oral mucosal barriers: A review with a focus on applications, culture conditions and barrier properties. Tissue Barriers 2018; 6:1479568. [PMID: 30252599 PMCID: PMC6389128 DOI: 10.1080/21688370.2018.1479568] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Understanding the function of oral mucosal epithelial barriers is essential for a plethora of research fields such as tumor biology, inflammation and infection diseases, microbiomics, pharmacology, drug delivery, dental and biomarker research. The barrier properties are comprised by a physical, a transport and a metabolic barrier, and all these barrier components play pivotal roles in the communication between saliva and blood. The sum of all epithelia of the oral cavity and salivary glands is defined as the blood-saliva barrier. The functionality of the barrier is regulated by its microenvironment and often altered during diseases. A huge array of cell culture models have been developed to mimic specific parts of the blood-saliva barrier, but no ultimate standard in vitro models have been established. This review provides a comprehensive overview about developed in vitro models of oral mucosal barriers, their applications, various cultivation protocols and corresponding barrier properties.
Collapse
Affiliation(s)
- Lisa Bierbaumer
- a Competence Unit Molecular Diagnostics, Center Health and Bioresources, Austrian Institute of Technology (AIT) GmbH , Vienna , Austria
| | - Uwe Yacine Schwarze
- b Department of Oral Biology , School of Dentistry, Medical University of Vienna , Vienna , Austria.,c Austrian Cluster for Tissue Regeneration , Vienna , Austria
| | - Reinhard Gruber
- b Department of Oral Biology , School of Dentistry, Medical University of Vienna , Vienna , Austria.,c Austrian Cluster for Tissue Regeneration , Vienna , Austria.,d Department of Periodontology , School of Dental Medicine, University of Bern , Bern , Switzerland
| | - Winfried Neuhaus
- a Competence Unit Molecular Diagnostics, Center Health and Bioresources, Austrian Institute of Technology (AIT) GmbH , Vienna , Austria
| |
Collapse
|
17
|
MIEN1 is tightly regulated by SINE Alu methylation in its promoter. Oncotarget 2018; 7:65307-65319. [PMID: 27589566 PMCID: PMC5323157 DOI: 10.18632/oncotarget.11675] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/18/2016] [Indexed: 01/26/2023] Open
Abstract
Migration and invasion enhancer 1 (MIEN1) is a novel gene involved in prostate cancer progression by enhancing prostate cancer cell migration and invasion. DNA methylation, an important epigenetic regulation, is one of the most widely altered mechanisms in prostate cancer. This phenomenon frames the basis to study the DNA methylation patterns in the promoter region of MIEN1. Bisulfite pyrosequencing demonstrates the MIEN1 promoter contains a short interspersed nuclear Alu element (SINE Alu) repeat sequence. Validation of methylation inhibition on MIEN1 was performed using nucleoside analogs and non-nucleoside inhibitors and resulted in an increase in both MIEN1 RNA and protein in normal cells. MIEN1 mRNA and protein increases upon inhibition of individual DNA methyltransferases using RNA interference technologies. Furthermore, dual luciferase reporter assays, in silico analysis, and chromatin immunoprecipitation assays identified a sequence upstream of the transcription start site that has a site for binding of the USF transcription factors. These results suggest the MIEN1 promoter has a SINE Alu region that is hypermethylated in normal cells leading to repression of the gene. In cancer, the hypomethylation of a part of this repeat, in addition to the binding of USF, results in MIEN1 expression.
Collapse
|
18
|
Kpetemey M, Chaudhary P, Van Treuren T, Vishwanatha JK. MIEN1 drives breast tumor cell migration by regulating cytoskeletal-focal adhesion dynamics. Oncotarget 2018; 7:54913-54924. [PMID: 27462783 PMCID: PMC5342390 DOI: 10.18632/oncotarget.10798] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
Migration and invasion enhancer 1 (MIEN1) is an important regulator of cell migration and invasion. MIEN1 overexpression represents an oncogenic event that promotes tumor cell dissemination and metastasis. The underlying mechanism by which MIEN1 regulates migration and invasion has yet to be deciphered. Here, we demonstrate that MIEN1 acts as a cytoskeletal-signaling adapter protein to drive breast cancer cell migration. MIEN1 localization is concentrated underneath the actin-enriched protrusive structures of the migrating breast cancer cells. Depletion of MIEN1 led to the loss of actin-protrusive structures whereas the over-expression of MIEN1 resulted in rich and thick membrane extensions. Knockdown of MIEN1 also decreased the cell-substratum adhesion, suggesting a role for MIEN1 in actin cytoskeletal dynamics. Our results show that MIEN1 supports the transition of G-actin to F-actin polymerization and stabilizes F-actin polymers. Additionally, MIEN1 promotes cellular adhesion and actin dynamics by inducing phosphorylation of FAK at Tyr-925 and reducing phosphorylation of cofilin at Ser-3, which results in breast cancer cell migration. Collectively, our data show that MIEN1 plays an essential role in maintaining the plasticity of the dynamic membrane-associated actin cytoskeleton, which leads to an increase in cell motility. Hence, targeting MIEN1 might represent a promising means to prevent breast tumor metastasis.
Collapse
Affiliation(s)
- Marilyne Kpetemey
- Department of Molecular and Medical Genetics, Institute for Cancer Research, and The Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Pankaj Chaudhary
- Department of Molecular and Medical Genetics, Institute for Cancer Research, and The Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Timothy Van Treuren
- Department of Molecular and Medical Genetics, Institute for Cancer Research, and The Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Jamboor K Vishwanatha
- Department of Molecular and Medical Genetics, Institute for Cancer Research, and The Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
19
|
Ren H, Qi Y, Yin X, Gao J. miR-136 targets MIEN1 and involves the metastasis of colon cancer by suppressing epithelial-to-mesenchymal transition. Onco Targets Ther 2017; 11:67-74. [PMID: 29339925 PMCID: PMC5744744 DOI: 10.2147/ott.s113359] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MIEN1 is a novel oncogene, and it involves tumor progression in various cancer types, including colon cancer. However, the definite molecular mechanisms of MIEN1 in colon cancer progression remain to be completely elucidated. In the present study, bioinformatics prediction showed that miR-136 could be an upstream regulator of MIEN1; a luciferase assay and Western blot assay revealed that miR-136 negatively regulates MIEN1 expression via directly targeting its 3'-untranslated region sequence. Moreover, a functional assay using wound healing and transwell invasion showed that overexpressed miR-136 inhibited cell migration and invasion, and overexpression of MIEN1 partly rescued the above-mentioned effects of miR-136 in colon cancer cells. Additionally, a clinical sample assay showed that miR-136 expression was generally downregulated in colon cancer tissue, which was inversely correlated with MIEN1 expression. Furthermore, we found that miR-136 suppressed the Akt/NF-κB signaling pathway and epithelial-to-mesenchymal transition in colon cancer. These results suggest that miR-136, as a tumor suppressor, acts in tumor metastasis by suppressing MIEN1 expression in colon cancer, providing a novel target for the treatment of colon cancer.
Collapse
Affiliation(s)
- Haipeng Ren
- Department of Internal Medicine of Oncology, People’s Hospital of Weifang, Weifang
| | - Yuanling Qi
- Department of Internal Medicine of Oncology, People’s Hospital of Weifang, Weifang
| | - Xiaoyan Yin
- Health and Family Planning Bureau of Weifang, Shouguang, People’s Republic of China
| | - Jianfeng Gao
- Department of Internal Medicine of Oncology, People’s Hospital of Weifang, Weifang
| |
Collapse
|
20
|
Yin K, Xu C, Zhao GH, Liu Y, Xiao T, Zhu S, Yan G. Construction of C35 gene bait recombinants and T47D cell cDNA library. Biosci Trends 2017; 11:550-556. [PMID: 29033401 DOI: 10.5582/bst.2017.01161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
C35 is a novel tumor biomarker associated with metastasis progression. To investigate the interaction factors of C35 in its high expressed breast cancer cell lines, we constructed bait recombinant plasmids of C35 gene and T47D cell cDNA library for yeast two-hybrid screening. Full length C35 sequences were subcloned using RT-PCR from cDNA template extracted from T47D cells. Based on functional domain analysis, the full-length C351-348bp was also truncated into two fragments C351-153bp and C35154-348bp to avoid auto-activation. The three kinds of C35 genes were successfully amplified and inserted into pGBKT7 to construct bait recombinant plasmids pGBKT7-C351-348bp, pGBKT7-C351-153bp and pGBKT7-C35154-348bp, then transformed into Y187 yeast cells by the lithium acetate method. Auto-activation and toxicity of C35 baits were detected using nutritional deficient medium and X-α-Gal assays. The T47D cell ds cDNA was generated by SMARTTM technology and the library was constructed using in vivo recombination-mediated cloning in the AH109 yeast strain using a pGADT7-Rec plasmid. The transformed Y187/pGBKT7-C351-348bp line was intensively inhibited while the truncated Y187/pGBKT7-C35 lines had no auto-activation and toxicity in yeast cells. The titer of established cDNA library was 2 × 107 pfu/mL with high transformation efficiency of 1.4 × 106, and the insert size of ds cDNA was distributed homogeneously between 0.5-2.0 kb. Our research generated a T47D cell cDNA library with high titer, and the constructed two C35 "baits" contained a respective functional immunoreceptor tyrosine based activation motif (ITAM) and the conserved last four amino acids Cys-Ile-Leu-Val (CILV) motif, and therefore laid a foundation for screening the C35 interaction factors in a BC cell line.
Collapse
Affiliation(s)
- Kun Yin
- Shandong Institute of Parasitical Disease, Shandong Academy of Medical Sciences
| | - Chao Xu
- Shandong Institute of Parasitical Disease, Shandong Academy of Medical Sciences
| | - Gui-Hua Zhao
- Shandong Institute of Parasitical Disease, Shandong Academy of Medical Sciences
| | - Ye Liu
- The First People's Hospital of Jining City
| | - Ting Xiao
- Shandong Institute of Parasitical Disease, Shandong Academy of Medical Sciences
| | - Song Zhu
- Shandong Institute of Parasitical Disease, Shandong Academy of Medical Sciences
| | - Ge Yan
- Shandong Institute of Parasitical Disease, Shandong Academy of Medical Sciences
| |
Collapse
|
21
|
Kwon MJ, Kim RN, Song K, Jeon S, Jeong HM, Kim JS, Han J, Hong S, Oh E, Choi JS, An J, Pollack JR, Choi YL, Park CK, Shin YK. Genes co-amplified with ERBB2 or MET as novel potential cancer-promoting genes in gastric cancer. Oncotarget 2017; 8:92209-92226. [PMID: 29190909 PMCID: PMC5696175 DOI: 10.18632/oncotarget.21150] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 08/27/2017] [Indexed: 12/18/2022] Open
Abstract
Gastric cancer (GC), one of the most common cancers worldwide, has a high mortality rate due to limited treatment options. Identifying novel and promising molecular targets is a major challenge that must be overcome if treatment of advanced GC is to be successful. Here, we used comparative genomic hybridization and gene expression microarrays to examine genome-wide DNA copy number alterations (CNAs) and global gene expression in 38 GC samples from old and young patients. We identified frequent CNAs, which included copy number gains on chromosomes 3q, 7p, 8q, 20p, and 20q and copy number losses on chromosomes 19p and 21p. The most frequently gained region was 7p21.1 (55%), whereas the most frequently deleted region was 21p11.1 (50%). Recurrent highly amplified regions 17q12 and 7q31.1-7q31.31 harbored two well-known oncogenes: ERBB2 and MET. Correlation analysis of CNAs and gene expression levels identified CAPZA2 (co-amplified with MET) and genes GRB7, MIEN1, PGAP3, and STARD3 (co-amplified with ERBB2) as potential candidate cancer-promoting genes (CPGs). Public dataset analysis confirmed co-amplification of these genes with MET or ERBB2 in GC tissue samples, and revealed that high expression (except for PGAP3) was significantly associated with shorter overall survival. Knockdown of these genes using small interfering RNA led to significant suppression of GC cell proliferation and migration. Reduced GC cell proliferation mediated by CAPZA2 knockdown was attributable to attenuated cell cycle progression and increased apoptosis. This study identified novel candidate CPGs co-amplified with MET or ERBB2, and suggests that they play a functional role in GC.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- College of Pharmacy, Kyungpook National University, Daegu, Korea.,Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Korea
| | - Ryong Nam Kim
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
| | - Kyoung Song
- R&D center, ABION Inc., Guro-gu, Seoul, Korea
| | - Sinyoung Jeon
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hae Min Jeong
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Joo Seok Kim
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Jinil Han
- Gencurix, Inc., Guro-gu, Seoul, Korea
| | - Sungyoul Hong
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Ensel Oh
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Sun Choi
- The Center for Anti-cancer Companion Diagnostics, Institutes of Entrepreneurial BioConvergence, Seoul National University, Seoul, Korea
| | - Jungsuk An
- Department of Pathology, Gachon University Gil Medical Center, Incheon, Korea
| | - Jonathan R Pollack
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yoon-La Choi
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Cheol-Keun Park
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea.,The Center for Anti-cancer Companion Diagnostics, Institutes of Entrepreneurial BioConvergence, Seoul National University, Seoul, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
22
|
Paquette M, Vilera-Perez LG, Beaudoin S, Ekindi-Ndongo N, Boudreaut PL, Bonin MA, Battista MC, Bentourkia M, Lopez AF, Lecomte R, Marsault E, Guérin B, Sabbagh R, Leyton JV. Targeting IL-5Rα with antibody-conjugates reveals a strategy for imaging and therapy for invasive bladder cancer. Oncoimmunology 2017; 6:e1331195. [PMID: 29123949 DOI: 10.1080/2162402x.2017.1331195] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/21/2022] Open
Abstract
Despite the high interest and concern due to an increasing incidence and death rate, patients who develop muscle invasive bladder cancer (MIBC) have few options available. However, the past decade has produced many candidate bladder tumor-specific markers but further development of these markers is still needed for creating effective targeted medications to solve this urgent need. Interleukin-5 receptor α-subunit (IL-5Rα) has recently been reported to be involved in MIBC progression. Thus, we aimed to validate IL-5Rα as a target for antibody-conjugates to better manage patients with MIBC. Patients were recruited and their tumors were processed for IL-5Rα immunohistochemical analysis. NOD/SCID mice were also heterotopically implanted with the human MIBC HT-1376 and HT-B9 cell lines and established xenografts immunohistochemically evaluated for IL-5Rα and compared against patient tumors. Using the mAb A14, an antibody-drug conjugate (ADC) and a radiolabeled immunoconjugate (RIC) were developed by conjugating to vinblastine and to the positron emitter copper-64 (64Cu), respectively. As a proof-of-concept for ADC and RIC efficacy, in vitro cytotoxicity and in vivo positron emission tomography (PET) imaging in tumor-bearing mice were performed, respectively. In addition, as rapid internalization and accumulation are important components for effective antibody-conjugates, we evaluated these aspects in response to IL-5 and 64Cu-A14 treatments. Our findings suggest that although IL-5Rα protein expression is preferentially increased in MIBC, it is rapid IL-5Rα-mediated internalization allowing vinblastine-A14 to have cytotoxic activity and 64Cu-A14 to detect MIBC tumors in vivo. This is the first report to elucidate the potential of IL-5Rα as an attractive MIBC target for antibody-conjugate applications.
Collapse
Affiliation(s)
- Michel Paquette
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences (FMHS), Centre hospitalier universitaire de Sherbrooke (CHUS), Université de Sherbrooke (UdeS), Sherbrooke, Québec, Canada
| | | | - Simon Beaudoin
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences (FMHS), Centre hospitalier universitaire de Sherbrooke (CHUS), Université de Sherbrooke (UdeS), Sherbrooke, Québec, Canada
| | | | - Pierre-Luc Boudreaut
- Department of Pharmacology and Physiology, FMHS, CHUS, UdeS, Sherbrooke, Québec, Canada
| | | | - Marie-Claude Battista
- Division of Endocrinology, Department of Medicine and Department of Obstetrics and Gynecology, FMHS, CHUS, Sherbrooke, Québec, Canada
| | - M'hamed Bentourkia
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences (FMHS), Centre hospitalier universitaire de Sherbrooke (CHUS), Université de Sherbrooke (UdeS), Sherbrooke, Québec, Canada.,Sherbrooke Molecular Imaging Centre (SMIC), Centre de recherche du CHUS (CRCHUS), Sherbrooke, Québec, Canada.,Sherbrooke Pharmacology Institute, Sherbrooke, Québec, Canada
| | - Angel F Lopez
- The Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Roger Lecomte
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences (FMHS), Centre hospitalier universitaire de Sherbrooke (CHUS), Université de Sherbrooke (UdeS), Sherbrooke, Québec, Canada.,Sherbrooke Molecular Imaging Centre (SMIC), Centre de recherche du CHUS (CRCHUS), Sherbrooke, Québec, Canada.,Sherbrooke Pharmacology Institute, Sherbrooke, Québec, Canada
| | - Eric Marsault
- Department of Pharmacology and Physiology, FMHS, CHUS, UdeS, Sherbrooke, Québec, Canada.,Sherbrooke Pharmacology Institute, Sherbrooke, Québec, Canada
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences (FMHS), Centre hospitalier universitaire de Sherbrooke (CHUS), Université de Sherbrooke (UdeS), Sherbrooke, Québec, Canada.,Sherbrooke Molecular Imaging Centre (SMIC), Centre de recherche du CHUS (CRCHUS), Sherbrooke, Québec, Canada.,Sherbrooke Pharmacology Institute, Sherbrooke, Québec, Canada
| | - Robert Sabbagh
- Department of Surgery, FMHS, CHUS, UdeS, Sherbrooke, Québec, Canada
| | - Jeffrey V Leyton
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences (FMHS), Centre hospitalier universitaire de Sherbrooke (CHUS), Université de Sherbrooke (UdeS), Sherbrooke, Québec, Canada.,Sherbrooke Molecular Imaging Centre (SMIC), Centre de recherche du CHUS (CRCHUS), Sherbrooke, Québec, Canada.,Sherbrooke Pharmacology Institute, Sherbrooke, Québec, Canada
| |
Collapse
|
23
|
Li D, Wei Y, Wang D, Gao H, Liu K. MicroRNA-26b suppresses the metastasis of non-small cell lung cancer by targeting MIEN1 via NF-κB/MMP-9/VEGF pathways. Biochem Biophys Res Commun 2016; 472:465-70. [DOI: 10.1016/j.bbrc.2016.01.163] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 01/25/2016] [Indexed: 12/15/2022]
|