1
|
Badve SS, Gökmen-Polar Y. Protein Profiling of Breast Cancer for Treatment Decision-Making. Am Soc Clin Oncol Educ Book 2022; 42:1-9. [PMID: 35580295 DOI: 10.1200/edbk_351207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The increasing use of neoadjuvant therapy has resulted in therapeutic decisions being made on the basis of diagnostic needle core biopsy. For many patients, this method might yield the only fragment of tumor available for biomarker analysis, necessitating judicious use. Many multiplex protein analytic methods have been developed that employ fluorescence or other tags to overcome the limitations of immunohistochemistry while still retaining the spatial annotation. Interpretation of the data can be difficult because of the limitations of the human eye. Computational deconvolution of the signals may be necessary for some of these methods to enable identification of cell-specific localization and coexpression of biomarkers. Herein, we present the different methods that are coming of age and their application in cancer research, with a focus on breast cancer. We also discuss the limitations, which include high costs and long turnaround times. The methods are also based on the premise that preanalytical factors will have identical impact on all proteins analyzed. There is a need to establish standards to normalize the data and enable cross-sample comparisons. In spite of these limitations, the multiplex technologies are extremely valuable discovery tools and can provide novel insights into the biology of cancer and mechanisms of drug resistance.
Collapse
Affiliation(s)
- Sunil S Badve
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Yesim Gökmen-Polar
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| |
Collapse
|
2
|
Li J, Bao S, Wang L, Wang R. CircZKSCAN1 Suppresses Hepatocellular Carcinoma Tumorigenesis by Regulating miR-873-5p/Downregulation of Deleted in Liver Cancer 1. Dig Dis Sci 2021; 66:4374-4383. [PMID: 33439397 DOI: 10.1007/s10620-020-06789-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third leading cause of cancer-associated mortality worldwide. CircZKSCAN1 (hsa_circ_0001727) was reported to be related to HCC development. The present study aims to elucidate the potential role and molecular mechanism of circZKSCAN1 in the regulation of HCC progression. METHODS CircZKSCAN1, miR-873-5p, and downregulation of deleted in liver cancer 1 (DLC1) in HCC tissues and cells were detected by RT-qPCR. Correlation between circZKSCAN1 expression and overall survival rate was measured by Kaplan-Meier survival analysis. The effects of circZKSCAN1, miR-873-5p, and DLC1 on proliferation, migration, and invasion were analyzed by CCK-8 and transwell assays, respectively. CyclinD1, Matrix metalloproteinase (MMP)-9, MMP-2, and DLC1 in HCC cells were detected by Western blot assay. The binding relationship between miR-873-5p and circZKSCAN1 or DLC1 was predicted by the Circinteractome or Starbase, and then confirmed by dual-luciferase reporter assays, respectively. Tumor volume and tumor weight were measured in vivo. RESULTS CircZKSCAN1 was downregulated in HCC tissues and cells. Kaplan-Meier survival analysis suggested that there was a positive correlation between circZKSCAN1 expression and overall survival rate. Functionally, circZKSCAN1 blocked proliferation, migration, and invasion of HCC cells. MiR-873-5p was a target miRNA of circZKSCAN1, and miR-873-5p directly bound with DLC1. Rescue experiments confirmed that miR-873-5p overexpression or DLC1 knockdown attenuated the suppressive effects of circZKSCAN1 on HCC tumor growth in vitro. Besides, circZKSCAN1 inhibited HCC cell growth in vivo. CONCLUSIONS This study firstly revealed that circZKSCAN1 curbed HCC progression via modulating miR-873-5p/DLC1 axis, providing a potential therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Jing Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Wannan Medical College, No. 10 Kangfu Road, Jinghu District, Wuhu City, 241000, Anhui Province, China.
| | - Siyang Bao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Wannan Medical College, No. 10 Kangfu Road, Jinghu District, Wuhu City, 241000, Anhui Province, China
| | - Linqi Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Wannan Medical College, No. 10 Kangfu Road, Jinghu District, Wuhu City, 241000, Anhui Province, China
| | - Ronglong Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Wannan Medical College, No. 10 Kangfu Road, Jinghu District, Wuhu City, 241000, Anhui Province, China
| |
Collapse
|
3
|
Neagu AN, Whitham D, Buonanno E, Jenkins A, Alexa-Stratulat T, Tamba BI, Darie CC. Proteomics and its applications in breast cancer. Am J Cancer Res 2021; 11:4006-4049. [PMID: 34659875 PMCID: PMC8493401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/05/2021] [Indexed: 06/13/2023] Open
Abstract
Breast cancer is an individually unique, multi-faceted and chameleonic disease, an eternal challenge for the new era of high-integrated precision diagnostic and personalized oncomedicine. Besides traditional single-omics fields (such as genomics, epigenomics, transcriptomics and metabolomics) and multi-omics contributions (proteogenomics, proteotranscriptomics or reproductomics), several new "-omics" approaches and exciting proteomics subfields are contributing to basic and advanced understanding of these "multiple diseases termed breast cancer": phenomics/cellomics, connectomics and interactomics, secretomics, matrisomics, exosomics, angiomics, chaperomics and epichaperomics, phosphoproteomics, ubiquitinomics, metalloproteomics, terminomics, degradomics and metadegradomics, adhesomics, stressomics, microbiomics, immunomics, salivaomics, materiomics and other biomics. Throughout the extremely complex neoplastic process, a Breast Cancer Cell Continuum Concept (BCCCC) has been modeled in this review as a spatio-temporal and holistic approach, as long as the breast cancer represents a complex cascade comprising successively integrated populations of heterogeneous tumor and cancer-associated cells, that reflect the carcinoma's progression from a "driving mutation" and formation of the breast primary tumor, toward the distant secondary tumors in different tissues and organs, via circulating tumor cell populations. This BCCCC is widely sustained by a Breast Cancer Proteomic Continuum Concept (BCPCC), where each phenotype of neoplastic and tumor-associated cells is characterized by a changing and adaptive proteomic profile detected in solid and liquid minimal invasive biopsies by complex proteomics approaches. Such a profile is created, beginning with the proteomic landscape of different neoplastic cell populations and cancer-associated cells, followed by subsequent analysis of protein biomarkers involved in epithelial-mesenchymal transition and intravasation, circulating tumor cell proteomics, and, finally, by protein biomarkers that highlight the extravasation and distant metastatic invasion. Proteomics technologies are producing important data in breast cancer diagnostic, prognostic, and predictive biomarkers discovery and validation, are detecting genetic aberrations at the proteome level, describing functional and regulatory pathways and emphasizing specific protein and peptide profiles in human tissues, biological fluids, cell lines and animal models. Also, proteomics can identify different breast cancer subtypes and specific protein and proteoform expression, can assess the efficacy of cancer therapies at cellular and tissular level and can even identify new therapeutic target proteins in clinical studies.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of IașiCarol I bvd. No. 22, Iași 700505, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Emma Buonanno
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Avalon Jenkins
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Teodora Alexa-Stratulat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and PharmacyIndependenței bvd. No. 16-18, Iași 700021, Romania
| | - Bogdan Ionel Tamba
- Advanced Center for Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and PharmacyMihail Kogălniceanu Street No. 9-13, Iași 700454, Romania
| | - Costel C Darie
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| |
Collapse
|
4
|
Tumor suppressor gene DLC1: Its modifications, interactive molecules, and potential prospects for clinical cancer application. Int J Biol Macromol 2021; 182:264-275. [PMID: 33836193 DOI: 10.1016/j.ijbiomac.2021.04.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022]
Abstract
Deleted in liver cancer 1 (DLC1) is a recognized tumor suppressor gene that negatively regulates Rho family proteins by hydrolyzing the active GTP-bound state to its inactive GDP-bound state. Active Rho proteins play a positive role in tumorigenesis. Numerous in vitro and in vivo experiments have shown that DLC1 is downregulated or inactivated in various solid tumors, which may be due to the following five reasons: genomic deletion, epigenetic modification and ubiquitin-dependent proteasomal degradation may cause DLC1 underexpression; phosphorylation at the post-translation level may cause DLC1 inactivation; and failure to localize at focal adhesions (FAs) may prevent DLC1 from exerting full activity. All of the causes could be attributed to molecular binding. Experimental evidence suggests that direct or indirect targeting of DLC1 is feasible for cancer treatment. Therefore, elucidating the interaction of DLC1 with its binding partners might provide novel targeted therapies for cancer. In this review, we summarized the binding partners of DLC1 at both the gene and protein levels and expounded a variety of anticancer drugs targeting DLC1 to provide information about DLC1 as a cancer diagnostic indicator or therapeutic target.
Collapse
|
5
|
Niu N, Ma X, Liu H, Zhao J, Lu C, Yang F, Qi W. DLC1 inhibits lung adenocarcinoma cell proliferation, migration and invasion via regulating MAPK signaling pathway. Exp Lung Res 2021; 47:173-182. [PMID: 33678109 DOI: 10.1080/01902148.2021.1885524] [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] [Indexed: 12/24/2022]
Abstract
Lung adenocarcinoma (LUAD), one of the most common cancers, is a major threat to people's health due to its high mortality, and the survival of most patients suffering LUAD remains poor. This study aimed to explore the mechanism of Deleted in Liver Cancer 1 (DLC1) as a tumor suppressor underlying the occurrence and progression of LUAD. As revealed by bioinformatics analysis and qRT-PCR, DLC1 was significantly down-regulated in LUAD tumor tissue and cells. A series of cellular experiments including CCK-8, wound healing and Transwell assays were performed to detect the effect of DLC1 on the biological function of LUAD cells. It was found that overexpressing DLC1 significantly inhibited LUAD cell proliferative, migratory and invasive abilities, while knockdown of DLC1 promoted these abilities. Gene Set Enrichment Analysis (GSEA) and dual-luciferase assay were used to explore the downstream signaling pathway of DLC1, finding that DLC1 could remarkably inhibit the activity of mitogen-activated protein kinase (MAPK) signaling pathway. Western blot implemented for MAPK signaling pathway-related proteins further identified that DLC1 restrained the activation of MAPK/ERK signaling pathway. Furthermore, rescue experiments suggested that DLC1 inhibited LUAD cell proliferation and invasion by suppressing the MAPK/ERK signaling pathway. Overall, our study discussed the DLC1-dependent mechanism involved in LUAD. We found that the up-regulation of DLC1 may inhibit the malignant progression of LUAD by suppressing MAPK signaling pathway, which supports the view that DLC1 may serve as a molecular target for the targeted therapy of LUAD patients.
Collapse
Affiliation(s)
- Niu Niu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Xingjie Ma
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Haitao Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Junjie Zhao
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Chao Lu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Fan Yang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Weibo Qi
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| |
Collapse
|
6
|
Liu H, Ma X, Niu N, Zhao J, Lu C, Yang F, Qi W. MIR-301b-3p Promotes Lung Adenocarcinoma Cell Proliferation, Migration and Invasion by Targeting DLC1. Technol Cancer Res Treat 2021; 20:1533033821990036. [PMID: 33754907 PMCID: PMC8093615 DOI: 10.1177/1533033821990036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/25/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND miR-301b-3p is reported in various human cancers for its abnormal expression, while the role and molecular mechanisms in lung adenocarcinoma (LUAD) remain unclear, and this is the focus of the present study. MATERIALS AND METHODS TCGA database was consulted to know gene expression in LUAD tissue. CCK-8, colony formation assay and Transwell assay were applied to identify the role of target genes in regulating LUAD cell biological properties. Bioinformatics analysis plus dual-luciferase assay were performed to validate the potential connection between genes. RESULTS miR-301b-3p and DLC1 were the target genes of this study and respectively differentially up-regulated and down-regulated in LUAD. Functional experiments indicated that miR-301b-3p contributed to cancer cell proliferation, migration and invasion, while this effect was reversed with overexpressed DLC1 which was identified as a direct target of and regulated by miR-301b-3p. CONCLUSIONS Collectively, miR-301b-3p was identified to actively function on LUAD malignant progression by suppressing DLC1 expression. This discovery provides a novel therapeutic strategy for LUAD patients, which helps improve the survival of patients.
Collapse
Affiliation(s)
- Haitao Liu
- Department of Cardiothoracic Surgery, The First Hospital of Jiaxing
(Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang China
| | - Xingjie Ma
- Department of Cardiothoracic Surgery, The First Hospital of Jiaxing
(Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang China
| | - Niu Niu
- Department of Cardiothoracic Surgery, The First Hospital of Jiaxing
(Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang China
| | - Junjie Zhao
- Department of Cardiothoracic Surgery, The First Hospital of Jiaxing
(Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang China
| | - Chao Lu
- Department of Cardiothoracic Surgery, The First Hospital of Jiaxing
(Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang China
| | - Fan Yang
- Department of Cardiothoracic Surgery, The First Hospital of Jiaxing
(Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang China
| | - Weibo Qi
- Department of Cardiothoracic Surgery, The First Hospital of Jiaxing
(Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang China
| |
Collapse
|
7
|
Flick KF, Yip-Schneider MT, Sublette CM, Simpson RE, Colgate CL, Wu H, Soufi M, Dewitt JM, Mosley AL, Ceppa EP, Zhang J, Schmidt CM. A Quantitative Global Proteomics Approach Identifies Candidate Urinary Biomarkers That Correlate With Intraductal Papillary Mucinous Neoplasm Dysplasia. Pancreas 2020; 49:1044-1051. [PMID: 32769857 DOI: 10.1097/mpa.0000000000001628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES A proteomic discovery study was performed to determine if urine possesses a unique biosignature that could form the basis for a noninvasive test able to predict intraductal papillary mucinous neoplasm (IPMN) dysplasia. METHODS Urine was collected from patients undergoing surgery for IPMN (72 low/moderate, 27 high-grade/invasive). Quantitative mass spectrometry-based proteomics was performed. Proteins of interest were identified by differential expression analysis followed by principal component analysis. RESULTS Proteomics identified greater than 4800 urinary proteins. Low/moderate and high-grade/invasive IPMN were distinguished by 188 proteins (P < 0.05). Following principal component analysis and heatmap visualization, vitamin D binding protein (DBP), apolipoprotein A1 (APOA1), and alpha-1 antitrypsin (A1AT) were selected. The proteomic abundance of DBP (median [interquartile range]) was significantly higher for high-grade/invasive than for low/moderate IPMN (219,735 [128,882-269,943] vs. 112,295 [77,905-180,773] normalized reporter ion intensity units; P = 0.001). Similarly, APOA1 was more abundant in the high-grade/invasive than low/moderate groups (235,420 [144,933-371,247] vs 150,095 [103,419-236,591]; P = 0.0007) as was A1AT (567,514 [358,544-774,801] vs 358,393 [260,850-477,882]; P = 0.0006). CONCLUSIONS Urinary DBP, APOA1, and A1AT represent potential biomarker candidates that may provide a noninvasive means of predicting IPMN dysplastic grade.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John M Dewitt
- Division of Gastroenterology, Department of Medicine
| | | | | | | | | |
Collapse
|
8
|
Zhang Y, Li G. A tumor suppressor DLC1: The functions and signal pathways. J Cell Physiol 2019; 235:4999-5007. [DOI: 10.1002/jcp.29402] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Yang Zhang
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life SciencesShandong Normal UniversityJinan China
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life SciencesShandong Normal UniversityJinan China
| |
Collapse
|
9
|
Ma X, Liu C, Gao C, Li J, Zhuang J, Liu L, Li H, Wang X, Zhang X, Dong S, Zhou C, Sun C. circRNA-associated ceRNA network construction reveals the circRNAs involved in the progression and prognosis of breast cancer. J Cell Physiol 2019; 235:3973-3983. [PMID: 31617204 DOI: 10.1002/jcp.29291] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/30/2019] [Indexed: 12/24/2022]
Abstract
Recently, increasing evidences show that circular RNAs (circRNAs) are important regulators of various diseases, especially cancer. However, the regulatory role and the potential mechanism of action of circRNAs in breast cancer remain largely unknown. In this study, weighted gene co-expression network analysis was conducted with the differentially expressed miRNAs and mRNAs in breast cancer from The Cancer Genome Atlas database to identify the key modules associated with the carcinogenesis of breast cancer. In the significant turquoise and brown modules, 22 miRNAs and 1877 mRNAs were identified, respectively. Then, We compared and predicted the target genes and performed survival analysis to identify the miRNAs and mRNAs related to the prognosis of breast cancer. A circRNA-related competitive endogenous RNA network was identified by database co-screening, and deleted in liver cancer 1 (DLC1) was identified as a key gene. Finally, to assess how genes in key modules and key genes contribute to the development of breast cancer, relevant pathway information was obtained through DAVID and Gene Set Enrichment Analysis. These data demonstrated that three circRNAs (hsa-circ-0083373, hsa-circ-0083374, and hsa-circ-0083375) that regulate DLC1 expression via hsa-mir-511 and are involved in the pathogenesis and development of breast cancer.
Collapse
Affiliation(s)
- Xiaoran Ma
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jie Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jing Zhuang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China.,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Lijuan Liu
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China
| | - Huayao Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xue Wang
- College of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xiaoming Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Shixia Dong
- Clinical Medical Colleges, Weifang Medical University, Weifang, Shandong, China
| | - Chao Zhou
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China
| | - Changgang Sun
- Department of Basic Medical Science, Qingdao University, Qingdao, 266071, China.,Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| |
Collapse
|
10
|
Chen WX, Lou M, Cheng L, Qian Q, Xu LY, Sun L, Zhu YL, Dai H. Bioinformatics analysis of potential therapeutic targets among ARHGAP genes in breast cancer. Oncol Lett 2019; 18:6017-6025. [PMID: 31788076 PMCID: PMC6864933 DOI: 10.3892/ol.2019.10949] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/16/2019] [Indexed: 12/19/2022] Open
Abstract
GTPase activating proteins (RhoGAPs) serve significant roles in multiple aspects of tumor biology. Genes encoding RhoGAPs (ARHGAP), which switch off Rho-like GTPases, are responsible for breast cancer biogenesis. However, the identification of suitable and novel biomarkers for precision treatment and prognosis remains challenging. The present study aimed to evaluate the expression of ARHGAP family genes in breast cancer and investigate the survival data using the Oncomine, Kaplan-Meier Plotter, bcGenExMiner and cBioPortal online databases. The results demonstrated low expression of ARHGAP6, 7, 10, 14, 19, 23 and 24 and high expression of ARHGAP9, 11, 15, 18 and 30 in patients with breast cancer compared with that in healthy individuals. The survival analysis revealed that low expression levels of ARHGAP6, 7 and 19 were associated with poor relapse-free survival (RFS) and overall survival (OS), whereas high expression levels of ARHGAP9, 15 and 30 were associated with preferable RFS and OS. Metastatic relapse data demonstrated that higher expression of ARHGAP9, 15, 18, 19, 25 and 30 were associated with better prognosis and increased expression of ARHGAP11A and 14 exerted negative effects on patient prognosis. The overlapping genes ARHGAP9, 15, 19 and 30 obtained from these bioinformatics analysis tools exhibited significant association with clinical parameters including age, the presence of estrogen receptor, progesterone receptor and epidermal growth factor receptor-2, Scarff-Bloom-Richardson grade and Nottingham prognostic index. In conclusion, bioinformatics analysis revealed that ARHGAP9, 15, 19 and 30, but not other ARHGAP family genes may be promising targets with prognostic value and biological function for precision treatment of breast cancer.
Collapse
Affiliation(s)
- Wei-Xian Chen
- Department of Breast Surgery, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China.,Department of Post-doctoral Working Station, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Ming Lou
- Department of Graduate School, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Lin Cheng
- Department of Breast Surgery, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Qi Qian
- Department of Breast Surgery, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Ling-Yun Xu
- Department of Breast Surgery, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Li Sun
- Department of Breast Surgery, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yu-Lan Zhu
- Department of Breast Surgery, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Hong Dai
- Department of Breast Surgery, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
11
|
Salimi K, Kip Ç, Çelikbıçak Ö, Usta DD, Pınar A, Salih B, Tuncel A. Ti (IV) attached‐phosphonic acid functionalized capillary monolith as a stationary phase for in‐syringe‐type fast and robust enrichment of phosphopeptides. Biomed Chromatogr 2019; 33:e4488. [DOI: 10.1002/bmc.4488] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/26/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Kouroush Salimi
- Department of Chemical Engineering, Faculty of Engineering and Natural SciencesAnkara Yildirim Beyazit University Ankara Turkey
| | - Çiğdem Kip
- Department of Chemical EngineeringHacettepe University Ankara Turkey
| | - Ömür Çelikbıçak
- Department of ChemistryHacettepe University Ankara Turkey
- Division of BioengineeringInstitute of Science Ankara Turkey
| | - Duygu Deniz Usta
- Department of Medical Biology and GeneticsGazi University Ankara Turkey
- Department of Medical Biologyİstanbul Medeniyet University İstanbul Turkey
| | - Aslı Pınar
- Department of Medical Biochemistry, Faculty of MedicineHacettepe University Ankara Turkey
| | - Bekir Salih
- Department of ChemistryHacettepe University Ankara Turkey
| | - Ali Tuncel
- Department of Chemical EngineeringHacettepe University Ankara Turkey
- Division of Nanotechnology and NanomedicineHacettepe University Ankara Turkey
| |
Collapse
|