1
|
Wei JB, Zeng XC, Ji KR, Zhang LY, Chen XM. Identification of Key Genes and Related Drugs of Adrenocortical Carcinoma by Integrated Bioinformatics Analysis. Horm Metab Res 2023. [PMID: 38109896 DOI: 10.1055/a-2209-0771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Adrenocortical carcinoma (ACC) is a malignant carcinoma with an extremely poor prognosis, and its pathogenesis remains to be understood to date, necessitating further investigation. This study aims to discover biomarkers and potential therapeutic agents for ACC through bioinformatics, enhancing clinical diagnosis and treatment strategies. Differentially expressed genes (DEGs) between ACC and normal adrenal cortex were screened out from the GSE19750 and GSE90713 datasets available in the GEO database. An online Venn diagram tool was utilized to identify the common DEGs between the two datasets. The identified DEGs were subjected to functional assessment, pathway enrichment, and identification of hub genes by performing the protein-protein interaction (PPI), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. The differences in the expressions of hub genes between ACC and normal adrenal cortex were validated at the GEPIA2 website, and the association of these genes with the overall patient survival was also assessed. Finally, on the QuartataWeb website, drugs related to the identified hub genes were determined. A total of 114 DEGs, 10 hub genes, and 69 known drugs that could interact with these genes were identified. The GO and KEGG analyses revealed a close association of the identified DEGs with cellular signal transduction. The 10 hub genes identified were overexpressed in ACC, in addition to being significantly associated with adverse prognosis in ACC. Three genes and the associated known drugs were identified as potential targets for ACC treatment.
Collapse
Affiliation(s)
- Jian-Bin Wei
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Xiao-Chun Zeng
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Kui-Rong Ji
- Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Ling-Yi Zhang
- Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Xiao-Min Chen
- Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
| |
Collapse
|
2
|
Hamidi AA, Taghehchian N, Zangouei AS, Akhlaghipour I, Maharati A, Basirat Z, Moghbeli M. Molecular mechanisms of microRNA-216a during tumor progression. Cancer Cell Int 2023; 23:19. [PMID: 36740668 PMCID: PMC9899407 DOI: 10.1186/s12935-023-02865-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) as the members of non-coding RNAs family are involved in post-transcriptional regulation by translational inhibiting or mRNA degradation. They have a critical role in regulation of cell proliferation and migration. MiRNAs aberrations have been reported in various cancers. Considering the importance of these factors in regulation of cellular processes and their high stability in body fluids, these factors can be suggested as suitable non-invasive markers for the cancer diagnosis. MiR-216a deregulation has been frequently reported in different cancers. Therefore, in the present review we discussed the molecular mechanisms of the miR-216a during tumor progression. It has been reported that miR-216a mainly functioned as a tumor suppressor through the regulation of signaling pathways and transcription factors. This review paves the way to suggest the miR-216a as a probable therapeutic and diagnostic target in cancer patients.
Collapse
Affiliation(s)
- Amir Abbas Hamidi
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- grid.411583.a0000 0001 2198 6209Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Basirat
- grid.411583.a0000 0001 2198 6209Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- grid.411583.a0000 0001 2198 6209Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran ,grid.411583.a0000 0001 2198 6209Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Exploring Potential Biomarkers, Ferroptosis Mechanisms, and Therapeutic Targets Associated with Cutaneous Squamous Cell Carcinoma via Integrated Transcriptomic Analysis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3524022. [PMID: 36247089 PMCID: PMC9553755 DOI: 10.1155/2022/3524022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/17/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022]
Abstract
Background Cutaneous squamous cell carcinoma (cSCC) is the leading cause of death in patients with nonmelanoma skin cancers (NMSC). However, the unclear pathogenesis of cSCC limits the application of molecular targeted therapy. Methods Three microarray datasets (GSE2503, GSE45164, and GSE66359) were downloaded from the Gene Expression Omnibus (GEO). After identifying the differentially expressed genes (DEGs) in tumor and nontumor tissues, five kinds of analyses, namely, functional annotation, protein-protein interaction (PPI) network, hub gene selection, TF-miRNA-mRNA regulatory network analysis, and ferroptosis mechanism, were performed. Results A total of 146 DEGs were identified with significant differences, including 113 upregulated genes and 33 downregulated genes. The enriched functions and pathways of the DEGs included microtubule-based movement, ATP binding, cell cycle, P53 signaling pathway, oocyte meiosis, and PLK1 signaling events. Nine hub genes were identified (CDK1, AURKA, RRM2, CENPE, CCNB1, KIAA0101, ZWINT, TOP2A, and ASPM). Finally, RRM2, AURKA, and SAT1 were identified as significant ferroptosis-related genes in cSCC. The differential expression of these genes has been verified in two other independent datasets. Conclusions By integrated bioinformatic analysis, the hub genes identified in this study elucidated the molecular mechanism of the pathogenesis and progression of cSCC and are expected to become future biomarkers or therapeutic targets.
Collapse
|
4
|
Wang K, Li J, Zhou B. KIAA0101 knockdown inhibits glioma progression and glycolysis by inactivating the PI3K/AKT/mTOR pathway. Metab Brain Dis 2022; 37:489-499. [PMID: 34792707 DOI: 10.1007/s11011-021-00863-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/28/2021] [Indexed: 11/25/2022]
Abstract
KIAA0101, a proliferating cell nuclear antigen (PCNA)-associated factor, is reported to be overexpressed and identified as an oncogene in several human malignancies. The purpose of this study is to determine the function and possible mechanism of KIAA0101 in glioma progression. KIAA0101 expression in glioma patients was analyzed by GSE50161 and GEPIA datasets. Kaplan-Meier survival analysis was used to evaluate the survival distributions. KIAA0101 expression in glioma cells were detected by qRT-PCR and western blot analyses. The function of KIAA0101 was investigated using MTT, flow cytometry, caspase-3 activity, and Transwell assays. Additionally, glycolytic flux was determined by measuring extracellular acidification rate (ECAR), glucose consumption, lactate production, and adenosine triphosphate (ATP) level. The changes of phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway were detected by western blot analysis. Results showed that KIAA0101 was upregulated in glioma tissues and cells. High KIAA0101 expression predicted a poor prognosis in glioma patients. KIAA0101 depletion impeded cell proliferation, migration, and invasion and triggered apoptosis in glioma cells. KIAA0101 silencing reduced the ECAR, glucose consumption, lactate production, and ATP level in glioma cells, suggesting that KIAA0101 knockdown inhibited glycolysis in glioma cells. Mechanistically, KIAA0101 knockdown inhibited the PI3K/AKT/mTOR pathway. In conclusion, KIAA0101 silencing inhibited glioma progression and glycolysis by inactivating the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Kai Wang
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, 223002, China
| | - Jinxiao Li
- Department of Neurosurgery, Xinyi People's Hospital, Xuzhou, 221400, China
| | - Botao Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Xuzhou Medical University, 32 Meijian Road, Xuzhou, 221006, China.
| |
Collapse
|
5
|
Liu LJ, Liao JM, Zhu F. Proliferating cell nuclear antigen clamp associated factor, a potential proto-oncogene with increased expression in malignant gastrointestinal tumors. World J Gastrointest Oncol 2021; 13:1425-1439. [PMID: 34721775 PMCID: PMC8529917 DOI: 10.4251/wjgo.v13.i10.1425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/11/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal (GI) cancers, including malignancies in the gastrointestinal tract and accessory organs of digestion, represent the leading cause of death worldwide due to the poor prognosis of most GI cancers. An investigation into the potential molecular targets of prediction, diagnosis, prognosis, and therapy in GI cancers is urgently required. Proliferating cell nuclear antigen (PCNA) clamp associated factor (PCLAF), which plays an essential role in cell proliferation, apoptosis, and cell cycle regulation by binding to PCNA, is a potential molecular target of GI cancers as it contributes to a series of malignant properties, including tumorigenesis, epithelial-mesenchymal transition, migration, and invasion. Furthermore, PCLAF is an underlying plasma prediction target in colorectal cancer and liver cancer. In addition to GI cancers, PCLAF is also involved in other types of cancers and autoimmune diseases. Several pivotal pathways, including the Rb/E2F pathway, NF-κB pathway, and p53-p21 cascade, are implicated in PCLAF-mediated diseases. PCLAF also contributes to some diseases through dysregulation of the p53 pathway, WNT signal pathway, MEK/ERK pathway, and PI3K/AKT/mTOR signal cascade. This review mainly describes in detail the role of PCLAF in physiological status and GI cancers. The signaling pathways involved in PCLAF are also summarized. Suppression of the interaction of PCLAF/PCNA or the expression of PCLAF might be potential biological therapeutic strategies for GI cancers.
Collapse
Affiliation(s)
- Li-Juan Liu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy & Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Jian-Ming Liao
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy & Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
- Department of Neurosurgery, Renmin Hospital, Wuhan University, Wuhan 430060, Hubei Province, China
| | - Fan Zhu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy & Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
6
|
Zhang J, Liu X, Zhou W, Lu S, Wu C, Wu Z, Liu R, Li X, Wu J, Liu Y, Guo S, Jia S, Zhang X, Wang M. Identification of Key Genes Associated With the Process of Hepatitis B Inflammation and Cancer Transformation by Integrated Bioinformatics Analysis. Front Genet 2021; 12:654517. [PMID: 34539726 PMCID: PMC8440810 DOI: 10.3389/fgene.2021.654517] [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: 01/16/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) has become the main cause of cancer death worldwide. More than half of hepatocellular carcinoma developed from hepatitis B virus infection (HBV). The purpose of this study is to find the key genes in the transformation process of liver inflammation and cancer and to inhibit the development of chronic inflammation and the transformation from disease to cancer. Methods Two groups of GEO data (including normal/HBV and HBV/HBV-HCC) were selected for differential expression analysis. The differential expression genes of HBV-HCC in TCGA were verified to coincide with the above genes to obtain overlapping genes. Then, functional enrichment analysis, modular analysis, and survival analysis were carried out on the key genes. Results We identified nine central genes (CDK1, MAD2L1, CCNA2, PTTG1, NEK2) that may be closely related to the transformation of hepatitis B. The survival and prognosis gene markers composed of PTTG1, MAD2L1, RRM2, TPX2, CDK1, NEK2, DEPDC1, and ZWINT were constructed, which performed well in predicting the overall survival rate. Conclusion The findings of this study have certain guiding significance for further research on the transformation of hepatitis B inflammatory cancer, inhibition of chronic inflammation, and molecular targeted therapy of cancer.
Collapse
Affiliation(s)
- Jingyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinkui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Zhou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhishan Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jiarui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Siyu Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shanshan Jia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaomeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Miaomiao Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
Zhang Y, Yao H, Li Y, Yang L, Zhang L, Chen J, Wang Y, Li X. Circular RNA TADA2A promotes proliferation and migration via modulating of miR‑638/KIAA0101 signal in non‑small cell lung cancer. Oncol Rep 2021; 46:201. [PMID: 34296306 PMCID: PMC8317161 DOI: 10.3892/or.2021.8152] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence indicates that circular (circ)RNAs exhibit complex functions in diverse malignant tumors, including non-small cell lung cancer (NSCLC). The role of the circRNA transcription adaptor 2A (circTADA2A) in NSCLC remains unclear. The expression, function and mechanism of circTADA2A in NSCLC development were investigated in the present study. The results revealed that circTADA2A was upregulated in NSCLC, and that knockdown of circTADA2A inhibited cell proliferation and migration in the NSCLC cell lines A549 and H1299. Functional assays demonstrated that circTADA2A promoted proliferation and migration via interacting with microRNA (miR)-638. Bioinformatics and reverse transcription-quantitative PCR assay confirmed that miR-638 was expressed at low levels in NSCLC. In addition, it was found that miR-638 served a tumor-suppressive role and suppressed proliferation and migration via PCNA clamp associated factor (KIAA0101) inhibition in A549 and H1299 cells. Lastly, it was verified that circTADA2A promoted cell proliferation and migration, at least partially, via miR-638/KIAA0101 signaling in A549 and H1299 cells. In summary, the present study showed that circTADA2A promoted NSCLC cell proliferation and migration via modulating miR-638/KIAA0101 signaling.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Radiation Oncology, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Hongmin Yao
- Department of Radiation Oncology, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Ying Li
- Department of Radiation Oncology, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Lu Yang
- First Department of Gastroenterology, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Liang Zhang
- Department of Breast Internal Medicine, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Jinxin Chen
- Department of Gynecological Oncology, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Yong Wang
- Central Laboratory, Central Hospital Affiliated to Shenyang Medical College, Dadong, Shenyang, Liaoning 110024, P.R. China
| | - Xia Li
- Department of Radiation Oncology, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Dadong, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
8
|
Lin P, Zhao Y, Li X, Liang Z. KIAA0101 in Malignant Pleural Mesothelioma: a Potential Diagnostic and Prognostic Marker. Comb Chem High Throughput Screen 2021; 25:1498-1506. [PMID: 34238152 DOI: 10.2174/1386207324666210707105634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/20/2021] [Accepted: 05/29/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Currently, there are no reliable diagnostic and prognostic markers for malignant pleural mesothelioma (MPM). The objective of this study was to identify hub genes that could be helpful for diagnosis and prognosis in MPM by using bioinformatics analysis. MATERIALS AND METHODS The gene expression profiles were downloaded from the Gene Expression Omnibus (GEO) database and The Cancer Genome Atlas (TCGA). Weighted gene co-expression network analysis (WGCNA), LASSO regression analysis, Cox regression analysis, and Gene Set Enrichment Analysis (GSEA) were performed to identify hub genes and their functions. RESULTS A total of 430 up-regulated and 867 downregulated genes in MPM were identified based on the GSE51024 dataset. According to the WGCNA analysis, differentially expressed genes were classified into 8 modules. Among them, the pink module was most closely associated with MPM. According to genes with GS > 0.8 and MM > 0.8, six genes were selected as candidate hub genes (NUSAP1, TOP2A, PLOD2, BUB1B, UHRF1, KIAA0101) in the pink module. In the LASSO model, three genes (NUSAP1, PLOD2, and KIAA0101) were identified with non-zero regression coefficients and were considered hub genes among the 6 candidates. The hub gene-based LASSO model can accurately distinguish MPM from controls (AUC = 0.98). Moreover, the high expression level of KIAA0101, PLOD2, and NUSAP1 were all associated with poor prognosis compared to the low level in Kaplan-Meier survival analyses. After further multivariate Cox analysis, only KIAA0101 (HR = 1.55, 95% CI = 1.05-2.29) was identified as an independent prognostic factor among these hub genes. Finally, GSEA revealed that high expression of KIAA0101 was closely associated with 10 signaling pathways. CONCLUSION Our study identified several hub genes relevant to MPM, including NUSAP1, PLOD2, and KIAA0101. Among these genes, KIAA0101 appears to be a useful diagnostic and prognostic biomarker for MPM, which may provide new clues for MPM diagnosis and therapy.
Collapse
Affiliation(s)
- Ping Lin
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuean Zhao
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoqian Li
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongan Liang
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
9
|
Kim MJ, Cervantes C, Jung YS, Zhang X, Zhang J, Lee SH, Jun S, Litovchick L, Wang W, Chen J, Fang B, Park JI. PAF remodels the DREAM complex to bypass cell quiescence and promote lung tumorigenesis. Mol Cell 2021; 81:1698-1714.e6. [PMID: 33626321 PMCID: PMC8052288 DOI: 10.1016/j.molcel.2021.02.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/15/2020] [Accepted: 01/29/2021] [Indexed: 01/01/2023]
Abstract
The DREAM complex orchestrates cell quiescence and the cell cycle. However, how the DREAM complex is deregulated in cancer remains elusive. Here, we report that PAF (PCLAF/KIAA0101) drives cell quiescence exit to promote lung tumorigenesis by remodeling the DREAM complex. PAF is highly expressed in lung adenocarcinoma (LUAD) and is associated with poor prognosis. Importantly, Paf knockout markedly suppressed LUAD development in mouse models. PAF depletion induced LUAD cell quiescence and growth arrest. PAF is required for the global expression of cell-cycle genes controlled by the repressive DREAM complex. Mechanistically, PAF inhibits DREAM complex formation by binding to RBBP4, a core DREAM subunit, leading to transactivation of DREAM target genes. Furthermore, pharmacological mimicking of PAF-depleted transcriptomes inhibited LUAD tumor growth. Our results unveil how the PAF-remodeled DREAM complex bypasses cell quiescence to promote lung tumorigenesis and suggest that the PAF-DREAM axis may be a therapeutic vulnerability in lung cancer.
Collapse
Affiliation(s)
- Moon Jong Kim
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher Cervantes
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Youn-Sang Jung
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Xiaoshan Zhang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sung Ho Lee
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Larisa Litovchick
- Department of Internal Medicine and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
10
|
Liu J, Gao L, Liao J, Yang J, Yuan F, Chen Q. Kiaa0101 serves as a prognostic marker and promotes invasion by regulating p38/snail1 pathway in glioma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:260. [PMID: 33708887 PMCID: PMC7940917 DOI: 10.21037/atm-20-3219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background Kiaa0101, a regulator of cell proliferation, is overexpressed in many malignant tumors. However, its role in promoting invasion of glioma is poorly understood. Here, we investigated the effects of Kiaa0101 on glioma invasion and elucidated the underlying mechanisms of action. Methods We analyzed Kiaa0101 expression using datasets from four public databases, namely TCGA, CGGA, Gravendeel and Rembrandt as well as experimentally on 123 glioma samples via western blot (WB), RT-PCR and immunohistochemistry (IHC). We further quantified migration and invasion using wound healing and transwell assays. WB, IHC and immunofluorescence (IF) were used to detect expression of invasion related markers. Moreover, we detected tumor invasion of glioma cells in vivo in 5-week-old Balb/c nude mice. Results Kiaa0101 was upregulated in glioma, relative to non-tumor brain tissues, with the expression increasing with increase in glioma grade. Kiaa0101 mRNA expression was especially enriched in isocitrate dehydrogenase (IDH)1 wild-type glioma. Kaplan-Meier analysis, based on the aforementioned datasets, revealed that high Kiaa0101 levels were significantly associated with worse overall survival. Besides, shRNA-mediated Kiaa0101 knockdown inhibited migration and invasion of glioma cells by reducing snail1 expression both in vitro and in vivo, whereas its upregulation enhanced malignant behaviors of these cells. Furthermore, Kiaa0101 regulated snail1 expression by activating the p38MAPK signaling pathway. Conclusions Our findings strongly indicate that Kiaa0101 is a prognostic biomarker for malignant tumors, and its inhibition may be an effective strategy for treating glioma.
Collapse
Affiliation(s)
- Junhui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lun Gao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jianmin Liao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ji'an Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fan'en Yuan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Hu S, Zeng W, Zhang W, Xu J, Yu D, Peng J, Wei Y. KIAA0101 as a new diagnostic and prognostic marker, and its correlation with gene regulatory networks and immune infiltrates in lung adenocarcinoma. Aging (Albany NY) 2020; 13:301-339. [PMID: 33231570 PMCID: PMC7835026 DOI: 10.18632/aging.104144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022]
Abstract
Proliferating cell nuclear antigen binding factor (encoded by KIAA0101/PCLAF) regulates DNA synthesis and cell cycle progression; however, whether the level of KIAA0101 mRNA in lung adenocarcinoma is related to prognosis and tumor immune infiltration is unknown. In patients with lung adenocarcinoma, the differential expression of KIAA0101 was analyzed using the Oncomine, GEPIA, and Ualcan databases. The prognosis of patients with different KIAA0101 expression levels was evaluated using databases such as Prognostan and GEPIA. Tumor immune infiltration associated with KIAA0101 was analyzed using TISIDB. Linkedmics was used to perform gene set enrichment analysis of KIAA0101. KIAA0101 expression in lung adenocarcinoma tissues was higher than that in normal lung tissues. Patients with lung adenocarcinoma with low KIAA0101 expression had a better prognosis than those with high KIAA0101 expression. We constructed the gene regulatory network of KIAA0101 in lung adenocarcinoma. KIAA0101 appeared to play an important role in the regulation of tumor immune infiltration and targeted therapy in lung adenocarcinoma. Thus, KIAA0101 mRNA levels correlated with the diagnosis, prognosis, immune infiltration, and targeted therapy in lung adenocarcinoma. These results provide new directions to develop diagnostic criteria, prognostic evaluation, immunotherapy, and targeted therapy for lung adenocarcinoma.
Collapse
Affiliation(s)
- Sheng Hu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weibiao Zeng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianjun Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dongliang Yu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jinhua Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
12
|
Cao H, Zheng J, Yao Y, Yang Q, Yan R, Sun W, Ruan K, Zhou J, Zhou J. Overexpression of KIAA0101 Promotes the Progression of Non-small Cell Lung Cancer. J Cancer 2020; 11:6663-6674. [PMID: 33046987 PMCID: PMC7545688 DOI: 10.7150/jca.45962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the leading cause of cancer related death worldwide, with a continue-rising incidence. The proliferating cell nuclear antigen binding protein KIAA0101 is highly expressed in various types of cancer, including non-small cell lung cancer (NSCLC). However, its biological role and underlying mechanisms in NSCLC remains unclear. We downloaded KIAA0101 mRNA and clinical data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), and verified the KIAA0101 expression by conducting experiments of immunochemistry (IHC), immunofluorescence (IF), quantitative real-time PCR (qRT-PCR) and western-blot. Functional experiments were performed to explore the biological roles in vitro and in vivo. The results showed that KIAA0101 was overexpressed in NSCLC tissues and cell lines. High KIAA0101 expression was associated with high T stage, nodal invasion, advanced tumor stage, and poor overall survival (P<0.01). The receiver operating characteristic (ROC) curves showed that KIAA0101 could distinguish NSCLC from paired normal tissues with statistical significance (AUC=0.969, P<0.001). The multivariate analysis revealed that KIAA0101 was an independent prognostic factor for overall survival (HR=1.249, 95% CI: 1.001-1.559, P=0.049). Furthermore, KIAA0101 knockdown induced G1 phase cell cycle arrest and inhibited NSCLC cell proliferation and migration. We also found that the depletion of KIAA0101 decreased tumor volume in nude mice. In summary, our findings suggested that KIAA0101 was a reliable diagnostic and prognostic factor in NSCLC, with potential to be a promising treatment target.
Collapse
Affiliation(s)
- He Cao
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yinan Yao
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qi Yang
- Department of Respiratory Disease, The First Affiliated Hospital of Jiaxing, Jiaxing University, Jiaxing 314000, China
| | - Runlan Yan
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Wenjia Sun
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Kexin Ruan
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
13
|
GATA3 and APOBEC3B are prognostic markers in adrenocortical carcinoma and APOBEC3B is directly transcriptionally regulated by GATA3. Oncotarget 2020; 11:3354-3370. [PMID: 32934779 PMCID: PMC7486697 DOI: 10.18632/oncotarget.27703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/14/2020] [Indexed: 02/01/2023] Open
Abstract
Recent evidence has implicated APOBEC3B (Apolipoprotein B mRNA editing enzyme catalytic subunit 3B) as a source of mutations in breast, bladder, cervical, lung, head, and neck cancers. However, the role of APOBEC3B in adrenocortical carcinoma (ACC) and the mechanisms through which its expression is regulated in cancer are not fully understood. Here, we report that APOBEC3B is overexpressed in ACC and it regulates cell proliferation by inducing S phase arrest. We show high APOBEC3B expression is associated with a higher copy number gain/loss at chromosome 4 and 8 and TP53 mutation rate in ACC. GATA3 was identified as a positive regulator of APOBEC3B expression and directly binds the APOBEC3B promoter region. Both GATA3 and APOBEC3B expression levels were associated with patient survival. Our study provides novel insights into the function and regulation of APOBEC3B expression in addition to its known mutagenic ability.
Collapse
|
14
|
Sun T, An Q, Yan R, Li K, Zhu K, Dang C, Yuan D. MicroRNA‑216a‑5p suppresses esophageal squamous cell carcinoma progression by targeting KIAA0101. Oncol Rep 2020; 44:1971-1984. [PMID: 32901882 PMCID: PMC7551273 DOI: 10.3892/or.2020.7751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
The KIAA0101 protein (also referred to as NS5ATP9 or Paf15) is overexpressed in esophageal squamous cell carcinoma (ESCC) and is associated with disease progression and poor patient survival, but how KIAA0101 expression is regulated remains unknown. The relationship between tumor miR-216a-5p expression and prognosis in patients with ESCC was revealed by survival analyses. Quantitative reverse-transcriptase PCR and western blot analysis were used to evaluate miR-216a-5p and KIAA0101 expression in human ESCC tissues and cell lines. The targeting of KIAA0101 by miR-216a-5p was verified by dual-luciferase reporter assays. The EC9706 and TE1 cell lines were transfected with miR-216a-5p mimics and inhibitor, or KIAA0101-specific shRNA and KIAA0101-expressing plasmids, in order to evaluate the effect of manipulating miR-216a-5p and KIAA0101 expression on ESCC cell proliferation, cell cycle progression, migration, and invasion. miR-216a-5p was lowly expressed and inversely correlated with KIAA0101 protein expression in ESCC tissues and cell lines. Lower miR-216a-5p expression was associated with worse prognosis in patients with ESCC. miR-216a-5p negatively regulated KIAA0101 expression by directly targeting the 3′-untranslated region of the KIAA0101 mRNA. Overexpression of miR-216a-5p suppressed the proliferation, migration, and invasion of the ESCC cell lines, whereas inhibition of miR-216a-5p had the opposite effects. Meanwhile, KIAA0101 promoted ESCC migration and invasion, and its overexpression abolished the antitumor effects of miR-216a-5p mimics. As a tumor suppressor, miR-216a-5p targets KIAA0101 to inhibit the proliferation, migration, and invasion of ESCC. Therefore, the miR-216a-5p/KIAA0101 axis may be a potential target for ESCC treatment.
Collapse
Affiliation(s)
- Tuanhe Sun
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Qi An
- Department of Rheumatism and Immunology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Rong Yan
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Kang Li
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Kun Zhu
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Chengxue Dang
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Dawei Yuan
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| |
Collapse
|
15
|
Zhao H, Chen M, Wang J, Cao G, Chen W, Xu J. PCNA-associated factor KIAA0101 transcriptionally induced by ELK1 controls cell proliferation and apoptosis in nasopharyngeal carcinoma: an integrated bioinformatics and experimental study. Aging (Albany NY) 2020; 12:5992-6017. [PMID: 32275642 PMCID: PMC7185143 DOI: 10.18632/aging.102991] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 03/09/2020] [Indexed: 12/16/2022]
Abstract
KIAA0101, previously identified as PCNA-associated factor, is overexpressed among almost majority of human cancers and has emerged as an important regulator of cancer progression; however, its function in human nasopharyngeal carcinoma (NPC) remain unknown. Integrated bioinformatics approaches were employed to determine the KIAA0101 expressions in the NPC samples. Lentiviral vectors carrying KIAA0101 shRNA were constructed and stable transfected cells were validated by qRT-PCR and western blot. Cellular functions were then evaluated by MTT, colony formation, Brdu staining, and flow cytometry. Mechanistic studies were systematically investigated by UCSC Genome Browser, GEO, UALCAN, QIAGEN, PROMO and JASPAR, ChIP, and the cBioPortal, et al. The results showed that KIAA0101 ranked top overexpressed gene lists in GSE6631 dataset. KIAA0101 was highly expressed in NPC tissues and cell lines. Furthermore, knockdown of KIAA0101 significantly inhibited cell proliferation and DNA replication, promoted apoptosis and cell cycle arrest in vitro. Meanwhile, the mechanistic study revealed that MAP kinase phosphorylation-dependent activation of ELK1 may enhance neighbor gene expressions of KIAA0101 and TRIP4 by binding both promotor regions in the NPC cells. Taken together, our findings indicate that overexpression of KIAA0101 activated by MAP kinase phosphorylation-dependent activation of ELK1 may play an important role in NPC progression.
Collapse
Affiliation(s)
- Hu Zhao
- Fujian Provincial Key Laboratory of Transplant Biology, Department of Urology, 900 Hospital of the Joint Logistics Team, Xiamen University, Fuzhou 350025, Fujian, P.R. China.,Office of Science Education, 900 Hospital of the Joint Logistics Team, Xiamen University, Fuzhou 350025, Fujian, P.R. China
| | - Miaosheng Chen
- Pathology Department, Longyan First Hospital Affiliated to Fujian Medical University, Longyan 364000, Fujian, P.R. China
| | - Jie Wang
- Fujian Provincial Key Laboratory of Transplant Biology, Department of Urology, 900 Hospital of the Joint Logistics Team, Xiamen University, Fuzhou 350025, Fujian, P.R. China
| | - Gang Cao
- Department of Oral and Maxillofacial Surgery, Medical School of Nanjing University, Nanjing 210002, Jiangsu, P.R. China
| | - Wei Chen
- Department of Oral and Maxillofacial Surgery, Medical School of Nanjing University, Nanjing 210002, Jiangsu, P.R. China
| | - Jinke Xu
- Department of Oral and Maxillofacial Surgery, Medical School of Nanjing University, Nanjing 210002, Jiangsu, P.R. China
| |
Collapse
|
16
|
Mitchell D, Chintala S, Fetcko K, Henriquez M, Tewari BN, Ahmed A, Bentley RT, Dey M. Common Molecular Alterations in Canine Oligodendroglioma and Human Malignant Gliomas and Potential Novel Therapeutic Targets. Front Oncol 2019; 9:780. [PMID: 31475119 PMCID: PMC6702544 DOI: 10.3389/fonc.2019.00780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/31/2019] [Indexed: 01/05/2023] Open
Abstract
Spontaneous canine (Canis lupus) oligodendroglioma (ODG) holds tremendous potential as an immunocompetent large animal model of human malignant gliomas (MG). However, the feasibility of utilizing this model in pre-clinical studies depends on a thorough understanding of the similarities and differences of the molecular pathways associated with gliomas between the two species. We have previously shown that canine ODG has an immune landscape and expression pattern of commonly described oncogenes similar to that of human MG. In the current study, we performed a comprehensive analysis of canine ODG RNAseq data from 4 dogs with ODG and 2 normal controls to identify highly dysregulated genes in canine tumors. We then evaluated the expression of these genes in human MG using Xena Browser, a publicly available database. STRING-database inquiry was used in order to determine the suggested protein associations of these differentially expressed genes as well as the dysregulated pathways commonly enriched by the protein products of these genes in both canine ODG and human MG. Our results revealed that 3,712 (23%) of the 15,895 differentially expressed genes demonstrated significant up- or downregulation (log2-fold change > 2.0). Of the 3,712 altered genes, ~50% were upregulated (n = 1858) and ~50% were downregulated (n = 1854). Most of these genes were also found to have altered expression in human MG. Protein association and pathway analysis revealed common pathways enriched by members of the up- and downregulated gene categories in both species. In summary, we demonstrate that a similar pattern of gene dysregulation characterizes both human MG and canine ODG and provide additional support for the use of the canine model in order to therapeutically target these common genes. The results of such therapeutic targeting in the canine model can serve to more accurately predict the efficacy of anti-glioma therapies in human patients.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sreenivasulu Chintala
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kaleigh Fetcko
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mario Henriquez
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brij N Tewari
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Atique Ahmed
- Department of Neurological Surgery, Northwestern University, Chicago, IL, United States
| | - R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Mahua Dey
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
17
|
Zhao H, Lai X, Zhang W, Zhu H, Zhang S, Wu W, Wang S, Tang M, Deng Z, Tan J. MiR-30a-5p frequently downregulated in prostate cancer inhibits cell proliferation via targeting PCLAF. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:278-289. [PMID: 30669858 DOI: 10.1080/21691401.2018.1553783] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) have vastly expanded our view of RNA world in intracellular signal regulating networks. Here, we functionally characterized a normally highly expressed miRNA, miR-30a-5p (MIMAT0000087), which exhibits downregulated expression profiles in prostate cancer samples. MiR-30a-5p knockdown and overexpression in PC-3 cell line alters cell proliferation supporting a tumour suppressor role. We also discovered that PCLAF is the direct target of miR-30a-5p. Notably, PC-3 cell proliferation is inhibited by miR-30a-5p/PCLAF axis. miR-30a-5p represents a novel molecule of functionally important miRNAs which may shed light on the novel therapeutic targets for prostate cancer.
Collapse
Affiliation(s)
- Hu Zhao
- a Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou Dongfang Hospital of Xiamen University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian, P.R. China.,b Fujian Provincial Key Laboratory of Transplant Biology, 900 Hospital of the Joint Logistics Team, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Xiaofeng Lai
- c Department of Clinical Genetics and Experimental Medicine, 900 Hospital of the Joint Logistics Team, Xiamen University School of Medicine, Fuzhou, Fujian, P. R. China
| | - Wei Zhang
- d Department of Urology, Bayi Hospital affiliated Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Hehuan Zhu
- a Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou Dongfang Hospital of Xiamen University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian, P.R. China
| | - Shenhang Zhang
- e ChinaFujian Key Laboratory of Exercise Rehabilitation, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P. R. China
| | - Weizhen Wu
- a Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou Dongfang Hospital of Xiamen University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian, P.R. China.,b Fujian Provincial Key Laboratory of Transplant Biology, 900 Hospital of the Joint Logistics Team, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Shuiliang Wang
- a Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou Dongfang Hospital of Xiamen University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian, P.R. China
| | - Minying Tang
- a Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou Dongfang Hospital of Xiamen University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian, P.R. China.,b Fujian Provincial Key Laboratory of Transplant Biology, 900 Hospital of the Joint Logistics Team, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Zhen Deng
- a Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou Dongfang Hospital of Xiamen University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian, P.R. China.,b Fujian Provincial Key Laboratory of Transplant Biology, 900 Hospital of the Joint Logistics Team, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Jianming Tan
- a Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou Dongfang Hospital of Xiamen University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian, P.R. China.,b Fujian Provincial Key Laboratory of Transplant Biology, 900 Hospital of the Joint Logistics Team, Fujian Medical University, Fuzhou, Fujian, P. R. China
| |
Collapse
|
18
|
Jain N, Roy J, Das B, Mallick B. miR-197-5p inhibits sarcomagenesis and induces cellular senescence via repression of KIAA0101. Mol Carcinog 2019; 58:1376-1388. [PMID: 31001891 DOI: 10.1002/mc.23021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/16/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022]
Abstract
The abnormal expressions of microRNAs (miRNAs) are known to be associated with various pathophysiological processes that lead to the development of a plethora of diseases including cancer. Among several miRNAs studied so far, miR-197 has been reported to play a vital role either as an oncogene or tumor suppressor in different cancers. However, its role in carcinogenesis of fibrosarcoma has not yet been elucidated. Therefore, the current study investigated the role of miR-197-5p, which is significantly downregulated in HT1080 fibrosarcoma cells compared to IMR90-tert fibroblast cells. The transient overexpression of miR-197-5p causes a significant decrease in viability and proliferation of fibrosarcoma cells in both concentration- and time-dependent manners. Interestingly, we did not observe any significant changes in cell cycle pattern or apoptotic cell populations, but rather noticed cellular senescence of fibrosarcoma cells upon overexpression of miR-197-5p. Further, this miRNA suppresses the metastatic properties, such as migration, invasion, and anchorage-independent growth of fibrosarcoma possibly through targeting KIAA0101, which is a proliferating cell nuclear antigen-associated factor and overexpressed in the malignancy. In nutshell, our result revealed that miR-197-5p acts as an oncosuppressor miRNA in fibrosarcoma through target regulation of KIAA0101, which can be exploited for developing RNA-based therapeutic strategies for the cure of this malignancy.
Collapse
Affiliation(s)
- Neha Jain
- RNAi and Functional Genomics Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Jyoti Roy
- RNAi and Functional Genomics Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Basudeb Das
- RNAi and Functional Genomics Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Bibekanand Mallick
- RNAi and Functional Genomics Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
19
|
Wang Z, Dang C, Yan R, Zhang H, Yuan D, Li K. [Screening of cell cycle-related genes regulated by KIAA0101 in gastric cancer]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 38:1151-1158. [PMID: 30377125 DOI: 10.3969/j.issn.1673-4254.2018.10.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To screen the genes related to cell cycle under regulation by KIAA0101 in gastric cancer. METHODS RT-PCR was used to detect the expression level of KIAA0101 gene in gastric cancer tissue and paired adjacent tissues. GO function enrichment analysis and KEGG pathway enrichment analysis were carried out using DAVID database. KEGG was used to map the pathways and the corresponding genes were analyzed. The list of genes associated with the KIAA0101 expression pattern was imported into TCGA cBioPortal to analyze the relationship between the interacting genes and generate a genetic topology map. The candidate genes were screened by RT-PCR. RESULTS The expression level of KIAA0101 mRNA was significantly higher in cancer tissues than in paired adjacent tissues (1.104 ± 0.379 vs 0.421 ± 0.172; P=0.0179). The system screened genes related with KIAA0101 from 478 tissues by pooled analysis of the expression intensity of all the gene probes. GO function analysis showed that the differential genes were mainly enriched in protein phosphorylation, RNA processing, cell cycle, DNA metabolism, protein transport, acetylation, apoptosis, proteolysis, and redox. The changes in the expression level of KIAA0101 mainly affect the gastric cancer-related pathways including cell cycle, spliceosome, DNA replication, and p53 signal transduction pathway. KEGG pathway maps and gene topology maps showed that the genes related to KIAA0101 (such as BUB1B, MAD2L1, CDC45, CDK1, CCNE1 and CCNB2) were also related to cell cycle. RT-PCR results confirmed significant increments of the expression levels of BUB1B, MAD2L, CDK1, CCNE1, and CCNB2 mRNA in gastric cancer tissues as compared with the paired adjacent gastric tissues (P < 0.05), but CDC45 mRNA did not show significant differential expression in gastric cancer tissues (P > 0.05). CONCLUSIONS KIAA0101 may affect cell cycle by regulating the expression of BUB1B, MAD2L1, CDK1, CCNE1 and CCNB2, and this finding may provide evidence for understanding how KIAA0101 affects cell cycle and for screening of tumor markers and selection of drug targets.
Collapse
Affiliation(s)
- Zhi Wang
- First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710061, China.,Shaanxi Tuberculosis Hospital, Xi'an 710100, China
| | - Chengxue Dang
- First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710061, China
| | - Rong Yan
- First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710061, China
| | - Hao Zhang
- First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710061, China
| | - Dawei Yuan
- First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710061, China
| | - Kang Li
- First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710061, China
| |
Collapse
|
20
|
Jin C, Liu Z, Li Y, Bu H, Wang Y, Xu Y, Qiu C, Yan S, Yuan C, Li R, Diao N, Zhang Z, Wang X, Liu L, Kong B. PCNA-associated factor P15PAF, targeted by FOXM1, predicts poor prognosis in high-grade serous ovarian cancer patients. Int J Cancer 2018; 143:2973-2984. [PMID: 30129654 DOI: 10.1002/ijc.31800] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/24/2018] [Accepted: 08/01/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Chengjuan Jin
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
- Department of Obstetrics and Gynecology; Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University; 650 XinSongjiang Road, Shanghai People's Republic of China
| | - Zhaojian Liu
- Department of Cell Biology; Shandong University School of Medicine; 44 Wenhua Xi Road, Jinan China
| | - Yingwei Li
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Hualei Bu
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Yu Wang
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Ying Xu
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Chunping Qiu
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Shi Yan
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Cunzhong Yuan
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Rongrong Li
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Nannan Diao
- Institute of Diagnostics, School of Medicine; Shandong University; 44 Wenhua Xi Road, Jinan China
| | - Zhiwei Zhang
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Xiangxiang Wang
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Lidong Liu
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| | - Beihua Kong
- Department of Obstetrics and Gynecology; Qilu Hospital, Shandong University; 107 Wenhua Xi Road, Jinan China
| |
Collapse
|
21
|
Yuan L, Qian G, Chen L, Wu CL, Dan HC, Xiao Y, Wang X. Co-expression Network Analysis of Biomarkers for Adrenocortical Carcinoma. Front Genet 2018; 9:328. [PMID: 30158955 PMCID: PMC6104177 DOI: 10.3389/fgene.2018.00328] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/31/2018] [Indexed: 01/08/2023] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy with a poor prognosis. And currently, there are no specific diagnostic biomarkers for ACC. In our study, we aimed to screen biomarkers for disease diagnosis, progression and prognosis. We firstly used the microarray data from public database Gene Expression Omnibus database to construct a weighted gene co-expression network, and then to identify gene modules associated with clinical features of ACC. Though this algorithm, a significant module with R2 = 0.64 (P = 9 × 10-5) was identified. Co-expression network and protein–protein interaction network were performed for screen the candidate hub genes. Checked by The Cancer Genome Atlas (TCGA) database, another independent dataset GSE19750, and GEPIA database, using one-way ANOVA, Pearson’s correlation, survival analysis, diagnostic capacity (ROC curve) and expression level revalidation, a total 12 real hub genes were identified. Gene ontology and KEGG pathway analysis of genes in the significant module revealed that the hub genes are significantly enriched in cell cycle regulation. Moreover, gene set enrichment analysis suggests that the samples with highly expressed hub genes are correlated with cell cycle. Taken together, our integrated analysis has identified 12 hub genes that are associated with the progression and prognosis of ACC; these hub genes might lead to poor outcomes by regulating the cell cycle.
Collapse
Affiliation(s)
- Lushun Yuan
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guofeng Qian
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Liang Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chin-Lee Wu
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Han C Dan
- Greenebaum Cancer Center, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Lu H, Han M, Yuan X, Tursun K, Zhang Y, Li Y, Li Z, Feng S, Zhou L, Pan Z, Wang Q, Han K, Liu S, Cheng J. Role of IL-6-mediated expression of NS5ATP9 in autophagy of liver cancer cells. J Cell Physiol 2018; 233:9312-9319. [PMID: 29227529 DOI: 10.1002/jcp.26343] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022]
Abstract
This study aimed to investigate the relationship between interleukin-6 (IL-6) and NS5ATP9 in autophagy of liver cancer cells. Autophagy is one of the important regulators of the replication of hepatitis C virus and the survival of tumors. IL-6 is a multifunctional cytokine that plays an important role in autophagy and development of many kinds of tumors. However, the role of IL-6 in autophagy has not been fully explored. A previous study had shown that a novel gene, NS5ATP9, could modulate autophagy. The present study demonstrated that human IL-6 recombinant protein induced autophagy of HepG2 cells. Conversely, autophagy decreased after IL-6 was silenced or neutralized with monoclonal antibody against human IL-6. In addition, NS5ATP9 was upregulated by IL-6 via nuclear factor-kappaB activation, as detected by Western blot. Further studies indicated that the induction of autophagy by IL-6 could be attenuated by silencing NS5ATP9. Interestingly, the expression of NS5ATP9, in turn, resulted in the upregulation of IL-6. In conclusion, IL-6 could induce autophagy by expressing NS5ATP9, while NS5ATP9 upregulated IL-6 levels in turn, which further induced autophagy.
Collapse
Affiliation(s)
- Hongping Lu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Ming Han
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China.,Peking University Ditan Teaching Hospital, Beijing, China
| | - Xiaoxue Yuan
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Kelbinur Tursun
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China.,The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Yu Zhang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Yaru Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Zhongshu Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Shenghu Feng
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China.,Peking University Ditan Teaching Hospital, Beijing, China
| | - Li Zhou
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China.,Peking University Ditan Teaching Hospital, Beijing, China
| | - Zhipeng Pan
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China.,Dalian University, Dalian, China
| | - Qi Wang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Kai Han
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Shunai Liu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Jun Cheng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China.,Peking University Ditan Teaching Hospital, Beijing, China
| |
Collapse
|
23
|
Kim MJ, Xia B, Suh HN, Lee SH, Jun S, Lien EM, Zhang J, Chen K, Park JI. PAF-Myc-Controlled Cell Stemness Is Required for Intestinal Regeneration and Tumorigenesis. Dev Cell 2018. [PMID: 29533773 DOI: 10.1016/j.devcel.2018.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The underlying mechanisms of how self-renewing cells are controlled in regenerating tissues and cancer remain ambiguous. PCNA-associated factor (PAF) modulates DNA repair via PCNA. Also, PAF hyperactivates Wnt/β-catenin signaling independently of PCNA interaction. We found that PAF is expressed in intestinal stem and progenitor cells (ISCs and IPCs) and markedly upregulated during intestinal regeneration and tumorigenesis. Whereas PAF is dispensable for intestinal homeostasis, upon radiation injury, genetic ablation of PAF impairs intestinal regeneration along with the severe loss of ISCs and Myc expression. Mechanistically, PAF conditionally occupies and transactivates the c-Myc promoter, which induces the expansion of ISCs/IPCs during intestinal regeneration. In mouse models, PAF knockout inhibits Apc inactivation-driven intestinal tumorigenesis with reduced tumor cell stemness and suppressed Wnt/β-catenin signaling activity, supported by transcriptome profiling. Collectively, our results unveil that the PAF-Myc signaling axis is indispensable for intestinal regeneration and tumorigenesis by positively regulating self-renewing cells.
Collapse
Affiliation(s)
- Moon Jong Kim
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bo Xia
- Center for Cardiovascular Regeneration, Houston Methodist Hospital Research Institute, Houston, TX, USA; Department of Cardiothoracic Surgery, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Han Na Suh
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sung Ho Lee
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Esther M Lien
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kaifu Chen
- Center for Cardiovascular Regeneration, Houston Methodist Hospital Research Institute, Houston, TX, USA; Department of Cardiothoracic Surgery, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and Health Science Center, Houston, TX 77030, USA; Program in Genetics and Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
24
|
Yan R, Zhu K, Dang C, Lan K, Wang H, Yuan D, Chen W, Meltzer SJ, Li K. Paf15 expression correlates with rectal cancer prognosis, cell proliferation and radiation response. Oncotarget 2018; 7:38750-38761. [PMID: 27246972 PMCID: PMC5122426 DOI: 10.18632/oncotarget.9606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 04/26/2016] [Indexed: 01/12/2023] Open
Abstract
Paf15, which participates in DNA repair, is overexpressed in numerous solid tumors. Blocking of Paf15 inhibits the growth of many types of cancer cells; while simultaneously enhancing cellular sensitivity to UV radiation. However, its expression and function in rectal cancer (RC) remain unknown. The current study was undertaken to assess the association of Paf15 expression with RC prognosis, as well as to explore the participation of Paf15 in the response of RC cells to irradiation. Increased Paf15 expression was observed in RC tissues and associated with pTNM stage and poor survival. In vitro, Paf15 induced increased RC cell proliferation while accelerating cell cycle progression, inhibiting cell death, and protecting against gamma radiation-induced DNA damage in RC cells. In conclusion, increased Paf15 expression is associated with increased RC proliferation, decreased patient survival, and a worse radiotherapeutic response.
Collapse
Affiliation(s)
- Rong Yan
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China.,Department of Medicine (GI Division) and Oncology, Johns Hopkins School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Kun Zhu
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Ke Lan
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Haonan Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Dawei Yuan
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Wei Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Stephen J Meltzer
- Department of Medicine (GI Division) and Oncology, Johns Hopkins School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Kang Li
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
25
|
Wierinckx A, Roche M, Legras-Lachuer C, Trouillas J, Raverot G, Lachuer J. MicroRNAs in pituitary tumors. Mol Cell Endocrinol 2017; 456:51-61. [PMID: 28089822 DOI: 10.1016/j.mce.2017.01.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/14/2016] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
Since the presence of microRNAs was first observed in normal pituitary, the majority of scientific publications addressing their role and the function of microRNAs in the pituitary have been based on pituitary tumor studies. In this review, we briefly describe the involvement of microRNAs in the synthesis of pituitary hormones and we present a comprehensive inventory of microRNA suppressors and inducers of pituitary tumors. Finally, we summarize the functional role of microRNAs in tumorigenesis, progression and aggressiveness of pituitary tumors, mechanisms contributing to the regulation (transcription factors, genomic modifications or epigenetic) or modulation (pharmacological treatment) of microRNAs in these tumors, and the interest of thoroughly studying the expression of miRNAs in body fluids.
Collapse
Affiliation(s)
- Anne Wierinckx
- Université Lyon 1, Université de Lyon, Lyon, France; Institut Universitaire de Technologie Lyon1, Université de Lyon, F-69622 Villeurbanne Cedex, France; INSERM U1052, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; CNRS UMR 5286, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, F-69373 Lyon Cedex 08, France.
| | | | - Catherine Legras-Lachuer
- Université Lyon 1, Université de Lyon, Lyon, France; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, F-69373 Lyon Cedex 08, France; ViroScan3D, F-01600 Trévoux, France; UMR CNRS 5557 UCBL USC INRA 1193 ENVL, Dynamique Microbienne et Transmission Virale, F-69100 Villeurbanne Cedex, France
| | - Jacqueline Trouillas
- Université Lyon 1, Université de Lyon, Lyon, France; Centre de Pathologie Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron F-69677, France
| | - Gérald Raverot
- Université Lyon 1, Université de Lyon, Lyon, France; INSERM U1052, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; CNRS UMR 5286, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; Fédération d'Endocrinologie, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, F-69677, France Université Lyon 1, Université de Lyon, Lyon, France
| | - Joël Lachuer
- Université Lyon 1, Université de Lyon, Lyon, France; Institut Universitaire de Technologie Lyon1, Université de Lyon, F-69622 Villeurbanne Cedex, France; INSERM U1052, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; CNRS UMR 5286, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, F-69373 Lyon Cedex 08, France
| |
Collapse
|
26
|
Newman R, Ahlfors H, Saveliev A, Galloway A, Hodson DJ, Williams R, Besra GS, Cook CN, Cunningham AF, Bell SE, Turner M. Maintenance of the marginal-zone B cell compartment specifically requires the RNA-binding protein ZFP36L1. Nat Immunol 2017; 18:683-693. [PMID: 28394372 PMCID: PMC5438597 DOI: 10.1038/ni.3724] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/09/2017] [Indexed: 12/15/2022]
Abstract
RNA-binding proteins of the ZFP36 family are best known for inhibiting the expression of cytokines through binding to AU-rich elements in the 3' untranslated region and promoting mRNA decay. Here we identified an indispensable role for ZFP36L1 as the regulator of a post-transcriptional hub that determined the identity of marginal-zone B cells by promoting their proper localization and survival. ZFP36L1 controlled a gene-expression program related to signaling, cell adhesion and locomotion; it achieved this in part by limiting expression of the transcription factors KLF2 and IRF8, which are known to enforce the follicular B cell phenotype. These mechanisms emphasize the importance of integrating transcriptional and post-transcriptional processes by RNA-binding proteins for maintaining cellular identity among closely related cell types.
Collapse
Affiliation(s)
- Rebecca Newman
- Laboratory of Lymphocyte Signalling and Development, The Babraham
Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
- Immune Receptor Activation Laboratory, The Francis Crick Institute,
1 Midland Road, London, NW1 1AT, United Kingdom
| | - Helena Ahlfors
- Laboratory of Lymphocyte Signalling and Development, The Babraham
Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| | - Alexander Saveliev
- Laboratory of Lymphocyte Signalling and Development, The Babraham
Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| | - Alison Galloway
- Laboratory of Lymphocyte Signalling and Development, The Babraham
Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| | - Daniel J Hodson
- Department of Haematology, University of Cambridge, The Clifford
Allbutt Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0AH,
United Kingdom
| | - Robert Williams
- Laboratory of Lymphocyte Signalling and Development, The Babraham
Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| | - Gurdyal S. Besra
- School of Biosciences, University of Birmingham, Birmingham, B15
2TT, United Kingdom
| | - Charlotte N Cook
- MRC Centre for Immune Regulation, School of Immunity and Infection,
University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Adam F Cunningham
- MRC Centre for Immune Regulation, School of Immunity and Infection,
University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Sarah E Bell
- Laboratory of Lymphocyte Signalling and Development, The Babraham
Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| | - Martin Turner
- Laboratory of Lymphocyte Signalling and Development, The Babraham
Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| |
Collapse
|
27
|
Samantarrai D, Mallick B. miR-429 inhibits metastasis by targeting KIAA0101 in Soft Tissue Sarcoma. Exp Cell Res 2017; 357:33-39. [PMID: 28432002 DOI: 10.1016/j.yexcr.2017.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 04/11/2017] [Accepted: 04/17/2017] [Indexed: 12/17/2022]
Abstract
Soft tissue sarcomas (STS) are a heterogeneous group of rare tumors with high metastatic potential. There being only a handful of publication on metastasis of STS, we investigated the miRNA mediated target gene regulations in modulating the metastatic processes in this cancer. In this study, we amalgamated gene and miRNA expression profiles of high-grade STS samples with miRNA target predictions and identified miR-429 targeting KIAA0101 as a novel pair, which remain unexplored in STS metastasis. We validated their expression in metastatic fibrosarcoma cell line, HT1080 and performed several functional assays using miRNA mimics and KIAA0101 over-expression vector to confirm their role in metastasis. We observed miR-429 is downregulated in HT1080 cells and acting as an anti-metastatic miRNA that inhibited proliferation, migration, anchorage independent growth and invasion by de-repressing KIAA0101. Moreover, the renilla luciferase reporter assay confirmed that miR-429 targets KIAA0101 by binding to its 3/UTR and influence its expression. Taken together, our work demonstrated miR-429 mediates deregulation of KIAA0101 by acting as an anti-metastatic miRNA that targets KIAA0101 pro-metastatic gene during metastasis of STS.
Collapse
Affiliation(s)
- Devyani Samantarrai
- RNAi and Functional Genomics Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| | - Bibekanand Mallick
- RNAi and Functional Genomics Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India.
| |
Collapse
|
28
|
Wang X, Wang SS, Zhou L, Yu L, Zhang LM. A network-pathway based module identification for predicting the prognosis of ovarian cancer patients. J Ovarian Res 2016; 9:73. [PMID: 27806724 PMCID: PMC5093979 DOI: 10.1186/s13048-016-0285-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 10/25/2016] [Indexed: 12/19/2022] Open
Abstract
Background This study aimed to screen multiple genes biomarkers based on gene expression data for predicting the survival of ovarian cancer patients. Methods Two microarray data of ovarian cancer samples were collected from The Cancer Genome Atlas (TCGA) database. The data in the training set were used to construct Reactome functional interactions network, which then underwent Markov clustering, supervised principal components, Cox proportional hazard model to screen significantly prognosis related modules. The distinguishing ability of each module for survival was further evaluated by the testing set. Gene Ontology (GO) functional and pathway annotations were performed to identify the roles of genes in each module for ovarian cancer. Results The network based approach identified two 7-gene functional interaction modules (31: DCLRE1A, EXO1, KIAA0101, KIN, PCNA, POLD3, POLD2; 35: DKK3, FABP3, IRF1, AIM2, GBP1, GBP2, IRF2) that are associated with prognosis of ovarian cancer patients. These network modules are related to DNA repair, replication, immune and cytokine mediated signaling pathways. Conclusions The two 7-gene expression signatures may be accurate predictors of clinical outcome in patients with ovarian cancer and has the potential to develop new therapeutic strategies for ovarian cancer patients.
Collapse
Affiliation(s)
- Xin Wang
- Department of Gynaecology and Obstetrics, The 306 Hospital of PLA, Beijing, 100037, China
| | - Shan-Shan Wang
- Outpatient Pharmacy, Outpatient Department, NO.16 Chengzhuang Fengtai Distinct, Beijing, 100071, China
| | - Lin Zhou
- Department of Gynaecology and Obstetrics, The 306 Hospital of PLA, Beijing, 100037, China
| | - Li Yu
- Department of Gynaecology and Obstetrics, The 306 Hospital of PLA, Beijing, 100037, China
| | - Lan-Mei Zhang
- Department of Gynaecology and Obstetrics, The 306 Hospital of PLA, Beijing, 100037, China.
| |
Collapse
|
29
|
Chen H, Xia B, Liu T, Lin M, Lou G. KIAA0101, a target gene of miR-429, enhances migration and chemoresistance of epithelial ovarian cancer cells. Cancer Cell Int 2016; 16:74. [PMID: 27708548 PMCID: PMC5037619 DOI: 10.1186/s12935-016-0353-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 09/23/2016] [Indexed: 12/21/2022] Open
Abstract
Background Ovarian cancer is a common type of gynecological malignancies, and is the fifth leading cause of cancer-related death in women in the United States. MiR-429 and KIAA0101 have been found to be involved in several human malignancies, respectively. However, the role of miR-429 and KIAA0101, and the correlation between them during development of epithelial ovarian cancer (EOC) remain to be investigated. Methods The expression of KIAA0101 in EOC tissues and cells was measured by Quantitative real-time PCR, western blot, and immunochemistry. Cell proliferation assay, colony formation assay, and transwell assay was performed to assess the role of miR-429 and KIAA0101 in regulation of proliferation, migration, and chemoresistance of EOC cells. Luciferase assay was used to test the Wnt/β-catenin signaling activity in response to depletion of KIAA0101 and overexpression of miR-429. Results We found that KIAA0101 was upregulated in metastatic EOC tissues, compared to primary EOC tissues, and KIAA0101 was required for the migration activity and chemoresistance of EOC cells by enhancing Wnt/β-catenin signaling. Furthermore, we revealed KIAA0101 is direct target of miR-429. Similar to knockdown of KIAA0101, overexpression of miR-429 reduced invasion and chemoresistance of EOC cells. Co-transfection of KIAA0101 partially abrogates the inhibitory effects on invasion and chemoresistance in EOC cells. Conclusions KIAA0101, a target gene of miR-429, was upregulated in the metastatic EOC tissues, and enhanced the migration activity and chemoresistance of EOC cells. Both miR-429 and KIAA0101 may represent the potential therapeutic targets of EOC.
Collapse
Affiliation(s)
- Hong Chen
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, 150 Haping Rd, Harbin, 150020 Heilongjiang China
| | - Bairong Xia
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, 150 Haping Rd, Harbin, 150020 Heilongjiang China
| | - Tianbo Liu
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, 150 Haping Rd, Harbin, 150020 Heilongjiang China
| | - Mei Lin
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, 150 Haping Rd, Harbin, 150020 Heilongjiang China
| | - Ge Lou
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, 150 Haping Rd, Harbin, 150020 Heilongjiang China
| |
Collapse
|
30
|
Alexandre KB, Mufhandu HT, London GM, Chakauya E, Khati M. Progress and Perspectives on HIV-1 microbicide development. Virology 2016; 497:69-80. [PMID: 27429040 DOI: 10.1016/j.virol.2016.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/01/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
The majority of HIV-1 infections occur via sexual intercourse. Women are the most affected by the epidemic, particularly in developing countries, due to their socio-economic dependence on men and the fact that they are often victims of gender based sexual violence. Despite significant efforts that resulted in the reduction of infection rates in some countries, there is still need for effective prevention methods against the virus. One of these methods for preventing sexual transmission in women is the use of microbicides. In this review we provide a summary of the progress made toward the discovery of affordable and effective HIV-1 microbicides and suggest future directions. We show that there is a wide range of compounds that have been proposed as potential microbicides. Although most of them have so far failed to show protection in humans, there are many promising ones currently in pre-clinical studies and in clinical trials.
Collapse
Affiliation(s)
- Kabamba B Alexandre
- Council for Scientific and Industrial Research, Pioneering Health Sciences Laboratory, Biosciences Unit, Pretoria, Gauteng, South Africa.
| | - Hazel T Mufhandu
- Council for Scientific and Industrial Research, Pioneering Health Sciences Laboratory, Biosciences Unit, Pretoria, Gauteng, South Africa
| | - Grace M London
- Department of Health Free State District Health Services and Health Programs, South Africa
| | - E Chakauya
- Council for Scientific and Industrial Research, Pioneering Health Sciences Laboratory, Biosciences Unit, Pretoria, Gauteng, South Africa
| | - M Khati
- Council for Scientific and Industrial Research, Pioneering Health Sciences Laboratory, Biosciences Unit, Pretoria, Gauteng, South Africa; University of Cape Town and Groote Schuur Hospital, Department of Medicine, Cape Town, South Africa
| |
Collapse
|
31
|
Quan M, Liu S, Wang Q, Li G, Zhang Y, Feng S, Liang J, Cheng J. NS5ATP9 Promotes Beclin 1-Dependent Starvation-Induced Autophagy of Hepatoblastoma Cells. J Cell Biochem 2016; 116:1574-82. [PMID: 25649430 DOI: 10.1002/jcb.25111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 01/23/2015] [Indexed: 12/16/2022]
Abstract
NS5ATP9, a gene up-regulated by NS5A, plays a crucial oncogenic role in several types of human tumours. However, the underlying mechanisms remain unclear. Autophagy, an evolutionarily conserved catabolic process, maintains cellular homeostasis under stress conditions, such as starvation, and plays a crucial role in tumour initiation and progression. Here, we report that NS5ATP9 mRNA and protein expression was up-regulated in starved HepG2 cells and that the up-regulated NS5ATP9 played a functional role in starvation-induced autophagy. Overexpression or silencing of this gene showed contrasting effects on Beclin 1 and on starvation-induced autophagy. Furthermore, NS5ATP9-mediated autophagy is required for promotion of tumour cell growth, and this effect could be inhibited with 3-methyladenine, chloroquine or by Beclin 1-silencing. Thus, the mechanism for NS5ATP9-promoted autophagy is Beclin 1-dependent in the condition of starvation, and for hepatoblastoma cell growth is also Beclin 1-dependent.
Collapse
Affiliation(s)
- Min Quan
- Department of General Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Shunai Liu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Qi Wang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Guoli Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Yu Zhang
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shenghu Feng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Jinqiu Liang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Jun Cheng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| |
Collapse
|
32
|
Abdelgawad IA, Radwan NH, Hassanein HR. KIAA0101 mRNA expression in the peripheral blood of hepatocellular carcinoma patients: Association with some clinicopathological features. Clin Biochem 2016; 49:787-91. [PMID: 26968109 DOI: 10.1016/j.clinbiochem.2015.12.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 12/30/2022]
Abstract
OBJECTIVES The development of hepatocellular carcinoma (HCC) is multi-factorial, multi-step and involving many genes. Recent studies have revealed the involvement of KIAA0101 in HCC development and progression. KIAA0101 is involved in the regulation of DNA repair, cell cycle progression, and cell proliferation. This study aims to elucidate the clinicopathological significance of KIAA0101 mRNA expression in the whole blood of HCC patients. DESIGN AND METHODS This study was conducted on 77 patients with proven HCC who presented to the outpatient clinic at the National Cancer Institute - Cairo University over a period of 8 consecutive months. Thirty patients with cirrhosis and forty apparently healthy volunteers were included as control groups. Detection of KIAA0101 mRNA was done on whole blood collected on EDTA for all patients and control subjects using real-time PCR. RESULTS KIAA0101 mRNA was over-expressed in the HCC group compared to the control groups. Overexpression of KIAA0101 mRNA was significantly associated with distant metastasis, advanced stage, high serum alkaline phosphatase and low serum albumin levels. Both sensitivity and specificity of KIAA0101 mRNA were higher than those of AFP and CEA. CONCLUSION Being associated with some of the prognostic factors of HCC which reflect tumor progression; as advanced stage, distant metastasis, hypoalbuminemia and elevated serum alkaline phosphatase, together with its relatively high diagnostic performance; KIAA0101 mRNA might be nominated to play a probable role in the diagnosis and prognosis prediction of HCC. Further studies on a wider scale are recommended to confirm these results.
Collapse
Affiliation(s)
- Iman A Abdelgawad
- Department of Clinical Pathology, NCI, Cairo University, National Cancer Institute, Fom Elkhalig, Kasr Elaini Street, Post Code No. 11796 Cairo, Egypt.
| | - Noha H Radwan
- Department of Clinical Pathology, NCI, Cairo University, National Cancer Institute, Fom Elkhalig, Kasr Elaini Street, Post Code No. 11796 Cairo, Egypt.
| | - Hala R Hassanein
- Department of Clinical Pathology, NCI, Cairo University, National Cancer Institute, Fom Elkhalig, Kasr Elaini Street, Post Code No. 11796 Cairo, Egypt.
| |
Collapse
|
33
|
NS5ATP9 suppresses activation of human hepatic stellate cells, possibly via inhibition of Smad3/phosphorylated-Smad3 expression. Inflammation 2015; 38:278-89. [PMID: 25300817 DOI: 10.1007/s10753-014-0031-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activation of hepatic stellate cell (HSC) is the central event in liver fibrosis. NS5ATP9 is related to many malignant tumors, but little is known about its function in HSC activation. The aim of this study is to investigate the role of NS5ATP9 in HSC activation in vitro. Genes related to liver fibrosis were detected after NS5ATP9 overexpression or silencing with or without transforming growth factor (TGF)-β1 stimulation in the human HSCs by real-time polymerase chain reaction and western blotting. Cell proliferation, migration, and apoptosis were tested, and the mechanisms underlying the effect of NS5ATP9 on HSC activation were studied. We showed that NS5ATP9 suppressed HSC activation and collagen production, with or without TGF-β1 induction. Also, NS5ATP9 inhibited cell proliferation and migration and promoted apoptosis. Furthermore, NS5ATP9 reduced basal and TGF-β1-mediated Smad3/phosphorylated-Smad3 expression. The existence of a physical complex between NS5ATP9 and Smad3 was illustrated. NS5ATP9 suppresses HSC activation, extracellular matrix production, and promotes apoptosis, in part through reducing Smad3/phosphorylated-Smad3 expression.
Collapse
|
34
|
Roche M, Wierinckx A, Croze S, Rey C, Legras-Lachuer C, Morel AP, Fusco A, Raverot G, Trouillas J, Lachuer J. Deregulation of miR-183 and KIAA0101 in Aggressive and Malignant Pituitary Tumors. Front Med (Lausanne) 2015; 2:54. [PMID: 26322309 PMCID: PMC4530307 DOI: 10.3389/fmed.2015.00054] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/22/2015] [Indexed: 01/15/2023] Open
Abstract
Changes in microRNAs (miRNAs) expression in many types of cancer suggest that they may be involved in crucial steps during tumor progression. Indeed, miRNAs deregulation has been described in pituitary tumorigenesis, but few studies have described their role in pituitary tumor progression toward aggressiveness and malignancy. To assess the role of miRNAs within the hierarchical cascade of events in prolactin (PRL) tumors during progression, we used an integrative genomic approach to associate clinical-pathological features, global miRNA expression, and transcriptomic profiles of the same human tumors. We describe the specific down-regulation of one principal miRNA, miR-183, in the 8 aggressive (A, grade 2b) compared to the 18 non-aggressive (NA, grades 1a, 2a) PRL tumors. We demonstrate that it acts as an anti-proliferative gene by directly targeting KIAA0101, which is involved in cell cycle activation and inhibition of p53-p21-mediated cell cycle arrest. Moreover, we show that miR-183 and KIAA0101 expression significantly correlate with the main markers of pituitary tumors aggressiveness, Ki-67 and p53. These results confirm the activation of proliferation in aggressive and malignant PRL tumors compared to non-aggressive ones. Importantly, these data also demonstrate the ability of such an integrative genomic strategy, applied in the same human tumors, to identify the molecular mechanisms responsible for tumoral progression even from a small cohort of patients.
Collapse
Affiliation(s)
- Magali Roche
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052/CNRS UMR 5286 Centre Léon Bérard , Lyon , France ; Université Lyon 1, Université de Lyon , Lyon , France ; ViroScan3D , Trévoux , France
| | - Anne Wierinckx
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052/CNRS UMR 5286 Centre Léon Bérard , Lyon , France ; Université Lyon 1, Université de Lyon , Lyon , France ; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7 , Lyon , France
| | - Séverine Croze
- Université Lyon 1, Université de Lyon , Lyon , France ; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7 , Lyon , France
| | - Catherine Rey
- ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7 , Lyon , France
| | - Catherine Legras-Lachuer
- Université Lyon 1, Université de Lyon , Lyon , France ; ViroScan3D , Trévoux , France ; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7 , Lyon , France ; UMR CNRS 5557 UCBL USC INRA 1193 ENVL, Dynamique Microbienne et Transmission Virale , Lyon , France
| | - Anne-Pierre Morel
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052/CNRS UMR 5286 Centre Léon Bérard , Lyon , France
| | - Alfredo Fusco
- Instituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università degli Studi di Napoli "Federico II" , Naples , Italy ; Instituto Nacional de Câncer (INCA) , Rio de Janeiro , Brazil
| | - Gérald Raverot
- Université Lyon 1, Université de Lyon , Lyon , France ; UMR 5292, Centre de Neurosciences de Lyon, CNRS, INSERM S1028 , Lyon , France ; Fédération d'Endocrinologie, Groupement Hospitalier Est, Hospices Civils de Lyon , Lyon , France
| | - Jacqueline Trouillas
- Université Lyon 1, Université de Lyon , Lyon , France ; UMR 5292, Centre de Neurosciences de Lyon, CNRS, INSERM S1028 , Lyon , France ; Centre de Pathologie Est, Groupement Hospitalier Est, Hospice Civils de Lyon , Bron , France
| | - Joel Lachuer
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052/CNRS UMR 5286 Centre Léon Bérard , Lyon , France ; Université Lyon 1, Université de Lyon , Lyon , France ; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7 , Lyon , France
| |
Collapse
|
35
|
Hladik F, Burgener A, Ballweber L, Gottardo R, Vojtech L, Fourati S, Dai JY, Cameron MJ, Strobl J, Hughes SM, Hoesley C, Andrew P, Johnson S, Piper J, Friend DR, Ball TB, Cranston RD, Mayer KH, McElrath MJ, McGowan I. Mucosal effects of tenofovir 1% gel. eLife 2015; 4. [PMID: 25647729 PMCID: PMC4391502 DOI: 10.7554/elife.04525] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/02/2015] [Indexed: 02/07/2023] Open
Abstract
Tenofovir gel is being evaluated for vaginal and rectal pre-exposure prophylaxis against HIV transmission. Because this is a new prevention strategy, we broadly assessed its effects on the mucosa. In MTN-007, a phase-1, randomized, double-blinded rectal microbicide trial, we used systems genomics/proteomics to determine the effect of tenofovir 1% gel, nonoxynol-9 2% gel, placebo gel or no treatment on rectal biopsies (15 subjects/arm). We also treated primary vaginal epithelial cells from four healthy women with tenofovir in vitro. After seven days of administration, tenofovir 1% gel had broad-ranging effects on the rectal mucosa, which were more pronounced than, but different from, those of the detergent nonoxynol-9. Tenofovir suppressed anti-inflammatory mediators, increased T cell densities, caused mitochondrial dysfunction, altered regulatory pathways of cell differentiation and survival, and stimulated epithelial cell proliferation. The breadth of mucosal changes induced by tenofovir indicates that its safety over longer-term topical use should be carefully monitored.
Collapse
Affiliation(s)
- Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, United States
| | - Adam Burgener
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Lamar Ballweber
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Raphael Gottardo
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, United States
| | - Slim Fourati
- Vaccine and Gene Therapy Institute of Florida, Port Saint Lucie, United States
| | - James Y Dai
- Department of Biostatistics, University of Washington, Seattle, United States
| | - Mark J Cameron
- Vaccine and Gene Therapy Institute of Florida, Port Saint Lucie, United States
| | - Johanna Strobl
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Sean M Hughes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, United States
| | - Craig Hoesley
- Department of Medicine, University of Alabama, Birmingham, United States
| | | | | | - Jeanna Piper
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - David R Friend
- CONRAD, Eastern Virginia Medical School, Arlington, United States
| | - T Blake Ball
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Ross D Cranston
- University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Kenneth H Mayer
- Fenway Health, Beth Israel Deaconess Hospital, Harvard Medical School, Boston, United States
| | - M Juliana McElrath
- Department of Medicine, University of Washington, Seattle, United States
| | - Ian McGowan
- University of Pittsburgh School of Medicine, Pittsburgh, United States
| |
Collapse
|
36
|
Butz H, Szabó PM, Nofech-Mozes R, Rotondo F, Kovacs K, Mirham L, Girgis H, Boles D, Patocs A, Yousef GM. Integrative bioinformatics analysis reveals new prognostic biomarkers of clear cell renal cell carcinoma. Clin Chem 2014; 60:1314-26. [PMID: 25139457 DOI: 10.1373/clinchem.2014.225854] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The outcome of clear cell renal cell carcinoma (ccRCC) is still unpredictable. Even with new targeted therapies, the average progression-free survival is dismal. Markers for early detection and progression could improve disease outcome. METHODS To identify efficient and hitherto unrecognized pathogenic factors of the disease, we performed a uniquely comprehensive pathway analysis and built a gene interaction network based on large publicly available data sets assembled from 28 publications, comprising a 3-prong approach with high-throughput mRNA, microRNA, and protein expression profiles of 593 ccRCC and 389 normal kidney samples. We validated our results on 2 different data sets of 882 ccRCC and 152 normal tissues. Functional analyses were done by proliferation, migration, and invasion assays following siRNA (small interfering RNA) knockdown. RESULTS After integration of multilevel data, we identified aryl-hydrocarbon receptor (AHR), grainyhead-like-2 (GRHL2), and KIAA0101 as new pathogenic factors. GRHL2 expression was associated with higher chances for disease relapse and retained prognostic utility after controlling for grade and stage [hazard ratio (HR), 3.47, P = 0.012]. Patients with KIAA0101-positive expression suffered worse disease-free survival (HR, 3.64, P < 0.001), and in multivariate analysis KIAA0101 retained its independent prognostic significance. Survival analysis showed that GRHL2- and KIAA0101-positive patients had significantly lower disease-free survival (P = 0.002 and P < 0.001). We also found that KIAA0101 silencing decreased kidney cancer cell migration and invasion in vitro. CONCLUSIONS Using an integrative system biology approach, we identified 3 novel factors as potential biomarkers (AHR, GRHL2 and KIAA0101) involved in ccRCC pathogenesis and not linked to kidney cancer before.
Collapse
Affiliation(s)
- Henriett Butz
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Peter M Szabó
- Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Roy Nofech-Mozes
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada
| | - Fabio Rotondo
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada
| | - Kalman Kovacs
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada
| | - Lorna Mirham
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada
| | - Hala Girgis
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada
| | - Dina Boles
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada
| | - Attila Patocs
- HAS-SE "Lendulet" Hereditary Endocrine Tumors Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - George M Yousef
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada;
| |
Collapse
|
37
|
Ou S, Liu GD, Tan Y, Zhou LS, Bai SR, Xue G, Li J, Yang Y, Cui J, Cheng JM, Gu JW. A time course study about gene expression of post-thermal injury with DNA microarray. Int J Dermatol 2014; 54:757-64. [PMID: 25069606 DOI: 10.1111/ijd.12534] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Burn injury is one of the most common and devastating forms of trauma in daily life. However, the exact sequence of events after burn injury remains unknown. OBJECTIVE This study aims to investigate gene expression alterations after burn injury. METHODS Microarray data set GSE8056 was downloaded from the Gene Expression Omnibus (GEO) database, including 12 samples, equally distributed in four groups: normal skin tissue as control and damaged tissues 1-3 days after burn (early period); 4-7 days after burn (middle period); and more than 7 days after burn (late period). Packages in R language were utilized to pre-process the data and filter out the differentially expressed genes (DEGs). Functional annotation of all three groups of DEGs was conducted by using clusters of orthologous groups analysis. The DEGs shared by all three groups were picked out and analyzed with STRING to set up a protein-protein interaction network. CFinder was chosen to implement module analysis, and expression analysis systematic explorer was then adopted to reveal the dysfunctional pathways for each module. RESULTS A total of 727, 782, and 445 DEGs were identified in the early, middle, and late period after burn, and 234 DEGs were identified as continually differentially expressed throughout all time periods, including genes encoding proinflammatory cytokines, such as interleukin (IL)-6, IL-8, and IL-1β, and genes associated with cell proliferation. Three modules associated with cell proliferation and inflammatory responses were generated from the protein-protein interaction network. CONCLUSION Our findings are beneficial for understanding the progression of the wound healing response after burn.
Collapse
Affiliation(s)
- Shan Ou
- Department of Anesthesiology, General Hospital of Chengdu Military Command, Chengdu, Sichuan.,Department of Neurosurgery, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| | - Guo-Dong Liu
- Department 8, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan Tan
- Office of Scientific Research and Training Administration, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| | - Le-Shun Zhou
- Department of Anesthesiology, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| | - Shu-Rong Bai
- Department of Anesthesiology, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| | - Gang Xue
- Department of Burns and Plastic Surgery, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| | - Jun Li
- Department of Anesthesiology, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| | - Yong Yang
- Department of Burns and Plastic Surgery, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| | - Jian Cui
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jing-Min Cheng
- Department of Neurosurgery, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| | - Jian-Wen Gu
- Department of Neurosurgery, General Hospital of Chengdu Military Command, Chengdu, Sichuan
| |
Collapse
|
38
|
Xie S, Chen Z, Wang Q, Song X, Zhang L. Comparisons of gene expression in normal, lesional, and non-lesional psoriatic skin using DNA microarray techniques. Int J Dermatol 2014; 53:1213-20. [PMID: 25041445 DOI: 10.1111/ijd.12476] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES This study was designed to explore the pathogenesis of psoriasis and to identify potential bio-targets. Genome array technology was used to analyze the gene expression profiles of lesional and non-lesional psoriatic skin samples and normal skin samples. METHODS Gene expression profile GSE14905 was downloaded from the Gene Expression Omnibus (GEO) database. This included skin biopsy samples from normal healthy donors (n = 21), lesional skin biopsy samples from psoriasis patients (n = 33), and non-lesional skin biopsy samples from psoriasis patients (n = 28). Differentially expressed genes (DEGs) were identified using the Limma package in R language. Functions of specific DEGs were predicted by Gene Ontology (GO) enrichment analysis. A protein-protein interaction network was constructed to display the interactions among common DEGs. Finally, DAVID and WebGestalt were used to achieve a functional analysis of common DEGs. RESULTS Totals of 1020, 562, and 643 genes, respectively, were identified as being differentially expressed in normal versus lesional, normal versus non-lesional, and lesional versus non-lesional samples. The specific DEGs in the three groups were enriched for several GO terms, including mitotic cell cycle, immune response, and response to organic matter. The 40 common DEGs in the three groups may be involved in the defense response pathway in the development of psoriasis. Furthermore, three genes (RGS1, SOCS3, and NAMPT) may play key roles in distinguishing lesional and non-lesional tissues from normal tissues, and 10 genes (PTRRC, ALDH1A3, SAMSA1, C15orf48, ZC3H12A, SOD2, IL8, LTF, RHCG, and IL7R) may play key roles in distinguishing non-lesional from normal and lesional samples. CONCLUSIONS These genes may be considered as potential diagnostic markers and targets of therapeutics in psoriasis.
Collapse
Affiliation(s)
- Shaoqiong Xie
- Traditional Chinese Medicine Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | | | | | | | | |
Collapse
|
39
|
NS5ATP9 mRNA levels in peripheral blood mononuclear cells predict prognosis in patients with gastric cancer. Med Oncol 2014; 31:106. [PMID: 24996800 DOI: 10.1007/s12032-014-0106-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 06/25/2014] [Indexed: 01/09/2023]
Abstract
The prognosis for gastric cancer is poor; therefore, there is urgent need for the development of novel, especially noninvasive surrogate biomarkers. NS5ATP9 is significantly overexpressed in gastric cancer tissues, and altered NS5ATP9 expression in human gastric cancer is associated with tumor recurrence. This study aimed to determine whether a correlation exists between NS5ATP9 mRNA levels in peripheral blood mononuclear cells (PBMCs) and gastric cancer prognosis in patients. NS5ATP9 mRNA levels were assessed by real-time RT-PCR in PBMC samples obtained from 207 gastric cancer patients, 47 patients with benign gastric lesions, and 78 healthy individuals. In addition, NS5ATP9 protein expression was evaluated by Western blot in PBMC samples from 30 gastric cancer patients and 30 healthy individuals, chosen randomly. Finally, clinicopathological data and survival of all gastric cancer patients were collected and analyzed. NS5ATP9 in PBMCs from gastric cancer patients was significantly upregulated both at mRNA and protein levels. Interestingly, NS5ATP9 mRNA overexpression in PBMCs was significantly associated with poor disease-free survival and overall survival of cancer patients, but not with tumor recurrence. Multivariate analysis showed NS5ATP9 mRNA levels and tumor lymph node metastasis were independent correlation with 3-year survival rate. Importantly, the diagnostic sensitivity and specificity (70.6 and 79.5 %, respectively) were higher for NS5ATP9 mRNA in PBMCs compared with the serum tumor markers CEA (45.0 and 84.6 %, respectively) and CA19-9 (61.5 and 69.2 %, respectively). These findings suggested that NS5ATP9 mRNA levels in PBMCs can be used for prognosis in gastric cancer patients.
Collapse
|
40
|
Quan M, Liu S, Li G, Wang Q, Zhang J, Zhang M, Li M, Gao P, Feng S, Cheng J. A functional role for NS5ATP9 in the induction of HCV NS5A-mediated autophagy. J Viral Hepat 2014; 21:405-15. [PMID: 24750205 DOI: 10.1111/jvh.12155] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autophagy has been shown to facilitate replication of hepatitis C virus (HCV); however, the mechanism by which HCV induces autophagy has not been fully established. NS5A, a nonstructural protein expressed by HCV, regulates numerous cellular pathways, including autophagy, by up-regulating Beclin 1; however, the underlying mechanism remains unclear. To obtain new insights into HCV-regulated autophagy, NS5ATP9 was overexpressed in HepG2 and L02 cells, resulting in up-regulation of endogenous Beclin 1 mRNA and protein levels, respectively. The luciferase-reporter assay results showed that both NS5A and NS5ATP9 could transactivate Beclin 1 promoter activity, but that NS5A could not transactivate the Beclin 1 promoter in NS5ATP9-silenced HepG2 and L02 cells. Up-regulation of Beclin 1 mRNA and protein expression by NS5A could also be attenuated by NS5ATP9 knock-down. Furthermore, the HepG2 and L02 cells that transiently overexpressed NS5ATP9 had enhanced accumulation of vacuoles carrying the autophagy marker LC3, consistent with the conversion of endogenous LC3-I to LC3-II. In contrast, the conversion of endogenous LC3-I to LC3-II could not be enhanced by NS5A in NS5ATP9-silenced HepG2 cells. These results highlight an important potential role for NS5ATP9 in HCV NS5A-induced hepatocyte autophagy.
Collapse
Affiliation(s)
- M Quan
- Beijing Ditan Hospital, Peking University Teaching Hospital, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Xie C, Yao M, Dong Q. Proliferating cell unclear antigen-associated factor (PAF15): A novel oncogene. Int J Biochem Cell Biol 2014; 50:127-31. [DOI: 10.1016/j.biocel.2014.02.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/21/2014] [Accepted: 02/28/2014] [Indexed: 01/30/2023]
|
42
|
Expression of KIAA0101 protein is associated with poor survival of esophageal cancer patients and resistance to cisplatin treatment in vitro. J Transl Med 2013; 93:1276-87. [PMID: 24145239 DOI: 10.1038/labinvest.2013.124] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 01/12/2023] Open
Abstract
The KIAA0101 protein is overexpressed in various human cancers, including esophageal cancer (EC). This study assessed the association of KIAA0101 protein with prognosis and resistance to chemotherapy in EC patients and then explored the role of KIAA0101 in EC cells in vitro. A total of 228 EC patients participated in the study. Tissue samples were collected for immunohistochemical analysis of KIAA0101 expression in tumor and normal tissues for association with clinicopathological and survival data. KIAA0101 cDNA or shRNA were transfected into EC cells for assessment of tumor cell viability, sensitivity to cisplatin treatment, and gene expression. Array-based comparative genomic hybridization (aCGH) was used to detect the changed copy-number alterations in cell lines expressing different levels of KIAA0101. Expression of KIAA0101 protein was upregulated in EC tissues, which was associated with pTNM stage, resistance to chemotherapy, tumor recurrence, and poor survival of EC patients. In vitro experiments showed that expression of KIAA0101 enhanced cell proliferation and upregulated cyclins A and B expression, leading to a reduced G1 phase of the cell cycle. KIAA0101 also induced resistance of EC Eca-109 and TE-1 cell lines to cisplatin treatment through a decrease in apoptosis. The aCGH data showed that levels of KIAA0101 expression altered chromosome stability, affecting genes that are associated with cancer progression. In conclusion, upregulated KIAA0101 expression is associated with EC progression, resistance to chemotherapy, and poor survival of the patients.
Collapse
|
43
|
Sang Y, Zang W, Yan Y, Liu Y, Fu Q, Wang K, Chen Y, Qi N. Study of differential effects of TGF-beta3/BMP2 on chondrogenesis in MSC cells by gene microarray data analysis. Mol Cell Biochem 2013; 385:191-8. [PMID: 24198161 DOI: 10.1007/s11010-013-1827-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/13/2013] [Indexed: 12/22/2022]
Abstract
In order to explore the differential effects of TGF-beta3 and BMP2 on chondrogenesis in mesenchymal stem cells (MSCs), the gene expression profiles of MSC treated with TGF-beta3 and BMP2 were subjected to systematic analysis on the gene and functional level. The gene expression profiles of MSCs (downloaded from Gene Expression Omnibus database) in the early and later stages, induced with TGF-beta2 and BMP2, were analyzed using packages within R software and the differentially expressed genes (DEGs) were screened. The DEGs both in the two experimental groups were subjected to Gene Ontology and pathway enrichment analysis. The protein-protein interaction (PPI) networks of the DEGs were constructed using cytoscape software. Among the DEGs, 1,194 genes were up-regulated and 580 genes were down-regulated. The up-regulated genes were mainly enriched in the TGF-beta and cell cycle signaling pathways and down-regulated genes were mainly enriched in the insulin-mediated signal pathway, metabolic pathway of fructose and mannose, and glycolysis/gluconeogenesis pathway. Based on the PPI network analysis, the genes of KIAA0101, NEDD4, and TINF2 were confirmed to be important on chondrogenesis. The analysis of DEGs both in TGF-beta3 and BMP2 treated MSCs indicates that the genes are mainly involved in the cell cycle and intracellular signaling pathways. Also the similar gene expression profile can be achieved by transcription factors or microRNAs (miR-199a-5p and miR-31-5p) based on our prediction, which can provide a new approach for the treatment of cartilage injury.
Collapse
Affiliation(s)
- Yunxia Sang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Jain M, Zhang L, He M, Zhang YQ, Patterson EE, Nilubol N, Shen M, Kebebew E. TOP2A is overexpressed and is a therapeutic target for adrenocortical carcinoma. Endocr Relat Cancer 2013; 20:361-70. [PMID: 23533247 PMCID: PMC4990817 DOI: 10.1530/erc-12-0403] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare but aggressive malignancy with no effective therapy for patients with unresectable disease. The aim of the current study was i) to evaluate TOP2A expression and function in human adrenocortical neoplasm and ACC cells and ii) to determine the anticancer activity of agents that target TOP2A. TOP2A mRNA and protein expression levels were evaluated in 112 adrenocortical tissue samples (21 normal adrenal cortex, 80 benign adrenocortical tumors, and 11 ACCs). In vitro siRNA knockdown of TOP2A in ACC cell lines (NCI-H295R and SW13) was used to determine its effect on cellular proliferation, cell cycle, anchorage-independent growth, and cellular invasion. We screened 14 TOP2A inhibitors for their anticancer activity in ACC cells. TOP2A mRNA and protein expression was significantly higher in ACC than in benign and normal adrenocortical tissue samples (P<0.05). Knockdown of TOP2A gene expression in ACC cell lines significantly decreased cell proliferation, anchorage-independent growth, and invasion (P<0.05). A screening assay in NCI-H295R cells showed that 11 of 14 TOP2A inhibitors had antiproliferative activity, 5 of the 14 TOP2A inhibitors had a higher antiproliferative activity than mitotane, and aclarubicin was the agent with the highest activity. Aclarubicin was validated to significantly decrease proliferation and tumor spheroid size in both NCI-H295R and SW13 ACC cell lines (P<0.05). Our results suggest that TOP2A is overexpressed in ACC, regulates cellular proliferation and invasion in ACC cells, and is an attractive target for ACC therapy. Of the TOP2A inhibitors screened, aclarubicin is a good candidate agent to test in future clinical trials for patients with locally advanced and metastatic ACC.
Collapse
Affiliation(s)
- Meenu Jain
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lisa Zhang
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mei He
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ya-Qin Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Erin E. Patterson
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Naris Nilubol
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Electron Kebebew
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
45
|
Wang Q, Wang Y, Li Y, Gao X, Liu S, Cheng J. NS5ATP9 contributes to inhibition of cell proliferation by hepatitis C virus (HCV) nonstructural protein 5A (NS5A) via MEK/extracellular signal regulated kinase (ERK) pathway. Int J Mol Sci 2013; 14:10539-51. [PMID: 23698777 PMCID: PMC3676852 DOI: 10.3390/ijms140510539] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/10/2013] [Accepted: 04/15/2013] [Indexed: 01/04/2023] Open
Abstract
Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) is a remarkable protein as it clearly plays multiple roles in mediating viral replication, host-cell interactions and viral pathogenesis. However, on the impact of cell growth, there have been different study results. NS5ATP9, also known as KIAA0101, p15PAF, L5, and OEACT-1, was first identified as a proliferating cell nuclear antigen-binding protein. Earlier studies have shown that NS5ATP9 might play an important role in HCV infection. The aim of this study is to investigate the function of NS5ATP9 on hepatocellular carcinoma (HCC) cell lines proliferation under HCV NS5A expression. The results showed that overexpression of NS5ATP9 inhibited the proliferation of Bel7402 cells, whereas knockdown of NS5ATP9 by interfering RNA promoted the growth of HepG2 cells. Under HCV NS5A expression, RNA interference (RNAi) targeting of NS5ATP9 could reverse the inhibition of HepG2 cell proliferation, suggesting that NS5ATP9 might be an anti-proliferation gene that plays an important role in the suppression of cell growth mediated by HCV NS5A via MEK/ERK signaling pathway. These findings might provide new insights into HCV NS5A and NS5ATP9.
Collapse
Affiliation(s)
- Qi Wang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; E-Mails: (Q.W.); (Y.W.); (X.G.); (S.L.)
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Yongsheng Wang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; E-Mails: (Q.W.); (Y.W.); (X.G.); (S.L.)
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Perking Union Medical College, Beijing 100050, China
| | - Yue Li
- Beijing Center for Physical and Chemical Analysis, Beijing 100094, China; E-Mail:
| | - Xuesong Gao
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; E-Mails: (Q.W.); (Y.W.); (X.G.); (S.L.)
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Shunai Liu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; E-Mails: (Q.W.); (Y.W.); (X.G.); (S.L.)
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Jun Cheng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; E-Mails: (Q.W.); (Y.W.); (X.G.); (S.L.)
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-10-8432-2006; Fax: +86-10-8432-2059
| |
Collapse
|
46
|
Chang CN, Feng MJ, Chen YL, Yuan RH, Jeng YM. p15(PAF) is an Rb/E2F-regulated S-phase protein essential for DNA synthesis and cell cycle progression. PLoS One 2013; 8:e61196. [PMID: 23593430 PMCID: PMC3617190 DOI: 10.1371/journal.pone.0061196] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 03/07/2013] [Indexed: 11/18/2022] Open
Abstract
The p15(PAF)/KIAA0101 protein is a proliferating cell nuclear antigen (PCNA)-associated protein overexpressed in multiple types of cancer. Attenuation of p15(PAF) expression leads to modifications in the DNA repair process, rendering cells more sensitive to ultraviolet-induced cell death. In this study, we identified that p15(PAF) expression peaks during the S phase of the cell cycle. We observed that p15(PAF) knockdown markedly inhibited cell proliferation, S-phase progression, and DNA synthesis. Depletion of p15(PAF) resulted in p21 upregulation, especially chromatin-bound p21. We further identified that the p15(PAF) promoter contains 3 E2F-binding motifs. Loss of Rb-mediated transcriptional repression resulted in upregulated p15(PAF) expression. Binding of E2F4 and E2F6 to the p15(PAF) promoter caused transcriptional repression. Overall, these results indicate that p15(PAF) is tightly regulated by the Rb/E2F complex. Loss of Rb/E2F-mediated repression during the G1/S transition phase leads to p15(PAF) upregulation, which facilitates DNA synthesis and S-phase progression.
Collapse
Affiliation(s)
- Chih-Ning Chang
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
| | - Mow-Jung Feng
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
| | - Yu-Ling Chen
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
| | - Ray-Hwang Yuan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Ming Jeng
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
47
|
Zhu K, Diao D, Dang C, Shi L, Wang J, Yan R, Yuan D, Li K. Elevated KIAA0101 expression is a marker of recurrence in human gastric cancer. Cancer Sci 2013; 104:353-9. [PMID: 23240630 DOI: 10.1111/cas.12083] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/29/2012] [Accepted: 12/15/2012] [Indexed: 12/18/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors with a high rate of recurrence, which results in surgery being unsuccessful. Therefore, it is important to find the reason for the surgery failing. The purpose of the present study was to investigate a factor related to recurrence. Expression of KIAA0101 was assessed in 61 paired human primary GC and non-cancerous gastric tissue using immunohistochemistry. After surgery, all 61 patients were followed regularly for more than 24 months or until death to analyze the 2-year survival rate and recurrence. After suppressing KIAA0101 by RNA interference in human GC cell lines, the cell viability was detected using MTT. We are first to find that KIAA0101 was elevated in GC tissues compared with paired non-cancerous gastric tissues. Immunohistochemical staining also revealed the predominant nuclear localization of KIAA0101 protein. Despite these findings, GC patients with elevated KIAA0101 expression levels exhibited a high recurrence and subsequently poor prognosis in the survival study. Also, cell viability was significantly inhibited after suppressing KIAA0101 in GC cells, suggesting that KIAA0101 might promote cancer cell proliferation. KIAA0101 is increased in human GC and is a marker of recurrence.
Collapse
Affiliation(s)
- Kun Zhu
- Department of Surgical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Liu L, Chen X, Xie S, Zhang C, Qiu Z, Zhu F. Variant 1 of KIAA0101, overexpressed in hepatocellular carcinoma, prevents doxorubicin-induced apoptosis by inhibiting p53 activation. Hepatology 2012; 56:1760-9. [PMID: 22576474 DOI: 10.1002/hep.25834] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 05/03/2012] [Indexed: 12/21/2022]
Abstract
UNLABELLED KIAA0101 overexpression was detected in numerous malignant solid tumors and involved in tumor progression; however, the correlation between KIAA0101 expression level and human hepatocellular carcinoma (HCC) was controversial. Our data revealed abnormal expression of the KIAA0101 transcript variant 1 (KIAA0101 tv1) at both messenger RNA and protein levels in HCC tissues and cell lines assessed by semiquantitative reverse-transcription polymerase chain reaction (RT-PCR), virtual northern blot, western blot, and immunohistochemical analysis, especially in stage 3-4 HCCs. NIH3T3 cells transfected with KIAA0101 tv1 induced colony formation in vitro and tumor xenorafts in vivo, implying the oncogenic potential of KIAA0101 tv1. Semiquantitative RT-PCR, real-time quantitative RT-PCR, and western blot analysis demonstrated that doxorubicin (Adriamycin, ADR) treatment down-regulated expression of the KIAA0101 tv1, whereas it increased the acetylation of the p53 protein. Additionally, KIAA0101 tv1 prevented cells from apoptosis caused by ADR through suppressing the acetylation of p53 at Lys382. Immunoprecipitation analysis and mammalian two-hybrid assay indicated that KIAA0101 tv1 bound to the transactivation region (1-42 amino acids) of p53 and strongly inhibits its transcriptional activity. Taken together, our data suggest that KIAA0101 tv1 played an important role in the late stage of metastatic HCC and prevented apoptosis after chemotherapeutic drug treatment through inhibiting the transcriptional activity of the p53 gene. CONCLUSION KIAA0101 tv1 may function as a regulator, promoting cell survival in HCC through regulating the function of p53. Suppression of the KIAA0101 tv1 function is likely to be a promising strategy to develop novel cancer therapeutic drugs.
Collapse
Affiliation(s)
- Lijuan Liu
- Department of Medical Microbiology, Wuhan University, Wuhan, People's Republic of China
| | | | | | | | | | | |
Collapse
|