1
|
Jia Y, Jia R, Chen Y, Lin X, Aishan N, li H, Wang L, Zhang X, Ruan J. The role of RNA binding proteins in cancer biology: A focus on FMRP. Genes Dis 2025; 12:101493. [PMID: 40271197 PMCID: PMC12017997 DOI: 10.1016/j.gendis.2024.101493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 04/25/2025] Open
Abstract
RNA-binding proteins (RBPs) act as crucial regulators of gene expression within cells, exerting precise control over processes such as RNA splicing, transport, localization, stability, and translation through their specific binding to RNA molecules. The diversity and complexity of RBPs are particularly significant in cancer biology, as they directly impact a multitude of RNA metabolic events closely associated with tumor initiation and progression. The fragile X mental retardation protein (FMRP), as a member of the RBP family, is central to the neurodevelopmental disorder fragile X syndrome and increasingly recognized in the modulation of cancer biology through its influence on RNA metabolism. The protein's versatility, stemming from its diverse RNA-binding domains, enables it to govern a wide array of transcript processing events. Modifications in FMRP's expression or localization have been associated with the regulation of mRNAs linked to various processes pertinent to cancer, including tumor proliferation, metastasis, epithelial-mesenchymal transition, cellular senescence, chemotherapy/radiotherapy resistance, and immunotherapy evasion. In this review, we emphasize recent findings and analyses that suggest contrasting functions of this protein family in tumorigenesis. Our knowledge of the proteins that are regulated by FMRP is rapidly growing, and this has led to the identification of multiple targets for therapeutic intervention of cancer, some of which have already moved into clinical trials or clinical practice.
Collapse
Affiliation(s)
- Yunlu Jia
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Ruyin Jia
- The Second School of Clinical Medicine of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yongxia Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Xuanyi Lin
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Nadire Aishan
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Han li
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Linbo Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Xiaochen Zhang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
2
|
Guo Q, Zhou Y, Ni H, Niu M, Xu S, Zheng L, Zhang W. The SIX2/PFN2 feedback loop promotes the stemness of gastric cancer cells. J Transl Med 2024; 22:832. [PMID: 39256760 PMCID: PMC11389068 DOI: 10.1186/s12967-024-05618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 08/18/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The roles of the transcriptional factor SIX2 have been identified in several tumors. However, its roles in gastric cancer (GC) progression have not yet been revealed. Our objective is to explore the impact and underlying mechanisms of SIX2 on the stemness of GC cells. METHODS Lentivirus infection was employed to establish stable expression SIX2 or PFN2 in GC cells. Gain- and loss-of-function experiments were conducted to detect changes of stemness markers, flow cytometry profiles, tumor spheroid formation, and tumor-initiating ability. ChIP, RNA-sequencing, tissue microarray, and bioinformatics analysis were performed to reveal the correlation between SIX2 and PFN2. The mechanisms underlying the SIX2/PFN2 loop-mediated effects were elucidated through tissue microarray analysis, RNA stability assay, IP-MS, Co-Immunoprecipitation, and inhibition of the JNK signaling pathway. RESULTS The stemness of GC cells was enhanced by SIX2. Mechanistically, SIX2 directly bound to PFN2's promoter and promoted PFN2 activity. PFN2, in turn, promoted the mRNA stability of SIX2 by recruiting RNA binding protein YBX-1, subsequently activating the downstream MAPK/JNK pathway. CONCLUSION This study unveils the roles of SIX2 in governing GC cell stemness, defining a novel SIX2/PFN2 regulatory loop responsible for this regulation. This suggests the potential of targeting the SIX2/PFN2 loop for GC treatment (Graphical Abstracts).
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, P. R. China
| | - Yi Zhou
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, P. R. China
| | - Haiwei Ni
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, P. R. China
| | - Miaomiao Niu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Drug Design and Optimization, Ministry of Education, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Shengtao Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, P. R. China
- Department of Hepatobiliary Surgery, The First People's Hospital of Kunshan, Suzhou, 215132, P. R. China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, P. R. China.
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, P. R. China.
| |
Collapse
|
3
|
Mi Y, Dong M, Zuo X, Cao Q, Gu X, Mi H, Xiao F. Genome-wide identification and analysis of epithelial-mesenchymal transition-related RNA-binding proteins and alternative splicing in a human breast cancer cell line. Sci Rep 2024; 14:11753. [PMID: 38783078 PMCID: PMC11116388 DOI: 10.1038/s41598-024-62681-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
Exploring the mechanism of breast cancer metastasis and searching for new drug therapeutic targets are still the focuses of current research. RNA-binding proteins (RBPs) may affect breast cancer metastasis by regulating alternative splicing (AS) during epithelial-mesenchymal transition (EMT). We hypothesised that during EMT development in breast cancer cells, the expression level of RBPs and the gene AS pattern in the cell were significantly changed on a genome-wide scale. Using GEO database, this study identified differentially expressed RBPs and differential AS events at different stages of EMT in breast cancer cells. By establishing the correlation network of differential RBPs and differential AS events, we found that RBM47, PCBP3, FRG1, SRP72, RBMS3 and other RBPs may regulate the AS of ITGA6, ADGRE5, TNC, COL6A3 and other cell adhesion genes. By further analysing above EMT-related RBPs and AS in breast cancer tissues in TCGA, it was found that the expression levels of ADAT2, C2orf15, SRP72, PAICS, RBMS3, APOBEC3G, NOA1, ACO1 and the AS of TNC and COL6A3 were significantly correlated with the prognosis of breast cancer patients. The expression levels of all 8 RBPs were significantly different in breast cancer tissues without metastasis compared with normal breast tissues. Conclusively, eight RBPs such as RBMS3 and AS of TNC and COL6A3 could be used as predictors of breast cancer prognosis. These findings need to be further explored as possible targets for breast cancer treatment.
Collapse
Affiliation(s)
- Yin Mi
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China.
| | - Meilian Dong
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China
| | - Xiaoxiao Zuo
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China
| | - Qinchen Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China
| | - Xiaobin Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China
| | - Hailong Mi
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fankai Xiao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Wang S, Sun H, Chen G, Wu C, Sun B, Lin J, Lin D, Zeng D, Lin B, Huang G, Lu X, Lin H, Liang Y. RNA-binding proteins in breast cancer: Biological implications and therapeutic opportunities. Crit Rev Oncol Hematol 2024; 195:104271. [PMID: 38272151 DOI: 10.1016/j.critrevonc.2024.104271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/05/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
RNA-binding proteins (RBPs) refer to a class of proteins that participate in alternative splicing, RNA stability, polyadenylation, localization and translation of RNAs, thus regulating gene expression in post-transcriptional manner. Dysregulation of RNA-RBP interaction contributes to various diseases, including cancer. In breast cancer, disorders in RBP expression and function influence the biological characteristics of tumor cells. Targeting RBPs has fostered the development of innovative therapies for breast cancer. However, the RBP-related mechanisms in breast cancer are not completely clear. In this review, we summarize the regulatory mechanisms of RBPs and their signaling crosstalk in breast cancer. Specifically, we emphasize the potential of certain RBPs as prognostic factors due to their effects on proliferation, invasion, apoptosis, and therapy resistance of breast cancer cells. Most importantly, we present a comprehensive overview of the latest RBP-related therapeutic strategies and novel therapeutic targets that have proven to be useful in the treatment of breast cancer.
Collapse
Affiliation(s)
- Shimeng Wang
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China
| | - Hexing Sun
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China
| | - Guanyuan Chen
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China
| | - Chengyu Wu
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China
| | - Bingmei Sun
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China
| | - Jiajia Lin
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China
| | - Danping Lin
- Department of Medical Oncology, Cancer Hospital of SUMC, Shantou 515000, China
| | - De Zeng
- Department of Medical Oncology, Cancer Hospital of SUMC, Shantou 515000, China
| | - Baohang Lin
- Department of Thyroid, Breast and Vascular Surgery, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Guan Huang
- Department of Pathology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xiaofeng Lu
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China
| | - Haoyu Lin
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China.
| | - Yuanke Liang
- Department of Thyroid and Breast Surgery, Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou 515041, China.
| |
Collapse
|
5
|
Zhang C, Xie L, Lin Z. Homeobox-D 1 and FTO form a transcriptional-epigenetic feedback loop to promote head and neck cancer proliferation. Cell Biol Int 2023; 47:1987-1998. [PMID: 37655555 DOI: 10.1002/cbin.12087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/29/2023] [Accepted: 08/20/2023] [Indexed: 09/02/2023]
Abstract
Transcription factors (TFs) and N6-methyladenosine (m6A) modifiers are involved in tumor progression through transcriptional regulation and posttranscriptional regulation of genes, respectively. However, the crosstalk and role of these two types of gene expression regulators in head and neck squamous cell carcinoma (HNSC) remains poorly understood. In this study, we demonstrate that the TF homeobox-D1 (HOXD1) and the m6A demethylase fat mass and obesity-associated protein (FTO) form a positive feedback loop to promote cell proliferation and survival in HNSC. Clinically, HOXD1 expression is dysregulated in multiple cancer types and is associated with worse prognosis in patients with HNSC, stomach adenocarcinoma, uterine corpus endometrial carcinoma, and pheochromocytoma and paraganglioma. Mechanistically, FTO is overexpressed in HNSC tumor samples and positively regulates HOXD1 expression in an m6A-dependent manner. Functionally, deficiency of HOXD1 relieved the resistance of HNSC cells to apoptosis and arrested tumor cells at the G0/G1 phase, thereby inhibiting cell growth, whereas overexpression of HOXD1 caused the opposite effect. Furthermore, HOXD1 activates the transcription of the oncogenic factor FTO by directly targeting its promoter. Downregulation of FTO mimicked the biological effect of HOXD1 knockdown on HNSC. Importantly, overexpression of HOXD1 significantly rescued the proliferation inhibition and apoptosis promotion of HNSC cells induced by deficiency of FTO. Together, our findings reveal HOXD1 as a novel prognostic predictor and a potential target for HNSC, providing mechanistic insights into the role of the HOXD1-FTO circuit in this cancer.
Collapse
Affiliation(s)
- Chunyan Zhang
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Linsen Xie
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Zhen Lin
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Feng Y, Zhu S, Liu T, Zhi G, Shao B, Liu J, Li B, Jiang C, Feng Q, Wu P, Wang D. Surmounting Cancer Drug Resistance: New Perspective on RNA-Binding Proteins. Pharmaceuticals (Basel) 2023; 16:1114. [PMID: 37631029 PMCID: PMC10458901 DOI: 10.3390/ph16081114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
RNA-binding proteins (RBPs), being pivotal elements in both physiological and pathological processes, possess the ability to directly impact RNA, thereby exerting a profound influence on cellular life. Furthermore, the dysregulation of RBPs not only induces alterations in the expression levels of genes associated with cancer but also impairs the occurrence of post-transcriptional regulatory mechanisms. Consequently, these circumstances can give rise to aberrations in cellular processes, ultimately resulting in alterations within the proteome. An aberrant proteome can disrupt the equilibrium between oncogenes and tumor suppressor genes, promoting cancer progression. Given their significant role in modulating gene expression and post-transcriptional regulation, directing therapeutic interventions towards RBPs represents a viable strategy for combating drug resistance in cancer treatment. RBPs possess significant potential as diagnostic and prognostic markers for diverse cancer types. Gaining comprehensive insights into the structure and functionality of RBPs, along with delving deeper into the molecular mechanisms underlying RBPs in tumor drug resistance, can enhance cancer treatment strategies and augment the prognostic outcomes for individuals afflicted with cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Peijie Wu
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.F.); (S.Z.); (T.L.); (G.Z.); (B.S.); (J.L.); (B.L.); (C.J.); (Q.F.)
| | - Dong Wang
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.F.); (S.Z.); (T.L.); (G.Z.); (B.S.); (J.L.); (B.L.); (C.J.); (Q.F.)
| |
Collapse
|
7
|
Tan T, Ma M, Xing S. Effect of circ_0000009 on lung adenocarcinoma progression by regulating PDZD2 in a ceRNA- and RBP- dependent manner. Gene 2023:147555. [PMID: 37307895 DOI: 10.1016/j.gene.2023.147555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Accumulating evidence now demonstrated that circular RNAs (circRNAs) are closely related to the pathogenesis of lung adenocarcinoma (LUAD). Through GEO2R online analysis, we screened hsa_circ_0000009 (circ_0000009) from the GEO database (GSE158695), and its expression in LUAD cancer tissues and cell lines was detected by RT-qPCR. The looping structure of circ_0000009 was tested by RNase R and actinomycin D experiments. The changes of proliferation were tested by CCK-8 or EdU assay. And the changes of apoptosis in A549 and H1299 cells were measured via flow cytometry. The A549 BALB/c tumor model was established to evaluate the influence of circ_0000009 on LUAD cell growth in vivo. In addition, experiments connected with ceRNA direction (mainly including bioinformatics prediction and luciferase reporter assay) and RNA Binding Protein (RBP) direction (mainly including RNA pull-down assay, RIP assay and mRNA stability assay) were further developed to reveal the regulatory mechanism of circ_0000009. The gene and protein levels in this project were assessed by RT-qPCR and western blotting analysis, respectively. The data manifested that circ_0000009 was in low expression in LUAD. The in vitro and in vivo experiments threw light on that overexpression of circ_0000009 dramatically suppressing LUAD tumorigenesis. Mechanistically, circ_0000009 promoted the expression of PDZD2 by sponging miR-154-3p. Furthermore, circ_0000009 stabilized PDZD2 by recruiting IGF2BP2. This study illustrated the mechanism that overexpressing of circ_0000009 suppressed LUAD progression by upregulating PDZD2 expression, providing an original treatment direction for LUAD.
Collapse
Affiliation(s)
- Tan Tan
- Department of Thoracic Surgery, Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Mingming Ma
- Department of Respiratory, the first People's Hospital of Xining, Xining, China
| | - Shigui Xing
- Department of Thoracic Surgery, Nanjing Gaochun People's Hospital, Nanjing, China.
| |
Collapse
|
8
|
Ou M, Chu Y, Zhang Q, Zhao H, Song Q. HOXA cluster antisense RNA 2 elevates KIAA1522 expression through microRNA-520d-3p and insulin like growth factor 2 mRNA binding protein 3 to promote the growth of vascular smooth muscle cells in thoracic aortic aneurysm. ESC Heart Fail 2022; 9:2955-2966. [PMID: 35730141 PMCID: PMC9715842 DOI: 10.1002/ehf2.13968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 03/15/2022] [Accepted: 04/27/2022] [Indexed: 11/12/2022] Open
Abstract
AIMS Recently, long non-coding RNAs (lncRNAs) have been revealed to mediate smooth muscle dysfunction in thoracic aortic aneurysm (TAA). LncRNA HOXA-AS2 has been proposed to engage in the regulation of diverse diseases. However, its function in TAA remains unknown. This study aimed to reveal the role and mechanism of HOXA-AS2 in VSMCs which were implicated in TAA formation. METHODS AND RESULTS RT-qPCR or western blot was performed to detect RNA or protein expression levels. The role of HOXA-AS2 in VSMCs was explored by functional assays. The relationship among HOXA-AS2/miR-520d-3p/KIAA1522/IGF2BP3 was analysed via mechanism assays. HOXA-AS2 was detected to have significantly high expression in TAA tissues and function as an oncogene to promote proliferation of VSMCs, while inhibiting cell apoptosis (Figure 1, **P < 0.01). HOXA-AS2 was unveiled to bind with miR-520d-3p (Figure 2, *P < 0.05, **P < 0.01) and further up-regulate KIAA1522 to facilitate the growth of VSMCs (Figure 3-4, *P < 0.05, **P < 0.01). HOXA-AS2 was also found to recruit IGF2BP3 to stabilize KIAA1522 mRNA (Figure 5, **P < 0.01). All data were displayed as mean ± standard deviation. CONCLUSIONS HOXA-AS2 up-regulates KIAA1522 through targeting miR-520d-3p/IGF2BP3 to drive VSMC growth in TAA.
Collapse
Affiliation(s)
- Minghui Ou
- Department of Vascular SurgeryQingdao Municipal HospitalQingdaoChina
| | - Yaonan Chu
- Department of Cardiovascular SurgeryShengli Oilfield Central HospitalDongyingChina
| | - Qian Zhang
- Department of ObstetricsQingdao Municipal HospitalQingdaoChina
| | - Huidong Zhao
- Department of ObstetricsQingdao Municipal HospitalQingdaoChina
| | - Qiang Song
- Department of Cardiovascular SurgeryShengli Oilfield Central HospitalDongyingChina
| |
Collapse
|
9
|
Wang L, Qiao C, Cao L, Cai S, Ma X, Song X, Jiang Q, Huang C, Wang J. Significance of HOXD transcription factors family in progression, migration and angiogenesis of cancer. Crit Rev Oncol Hematol 2022; 179:103809. [PMID: 36108961 DOI: 10.1016/j.critrevonc.2022.103809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 10/31/2022] Open
Abstract
The transcription factors (TFs) of the HOX family play significant roles during early embryonic development and cellular processes. They also play a key role in tumorigenesis as tumor oncogenes or suppressors. Furthermore, TFs of the HOXD geFIne cluster affect proliferation, migration, and invasion of tumors. Consequently, dysregulated activity of HOXD TFs has been linked to clinicopathological characteristics of cancer. HOXD TFs are regulated by non-coding RNAs and methylation of DNA on promoter and enhancer regions. In addition, HOXD genes modulate the biological function of cancer cells via the MEK and AKT signaling pathways, thus, making HOXD TFs, a suitable molecular marker for cancer prognosis and therapy. In this review, we summarized the roles of HOXD TFs in different cancers and highlighted its potential as a diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Lumin Wang
- Gastroenterology department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Institute of precision medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Chenyang Qiao
- Gastroenterology department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Li Cao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Shuang Cai
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Xiaoping Ma
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Xinqiu Song
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an, Shaanxi, PR China
| | - Qiuyu Jiang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China.
| | - Jinhai Wang
- Gastroenterology department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Institute of precision medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
10
|
Chen Y, Qin H, Zheng L. Research progress on RNA-binding proteins in breast cancer. Front Oncol 2022; 12:974523. [PMID: 36059653 PMCID: PMC9433872 DOI: 10.3389/fonc.2022.974523] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is the most common malignancy in women and has a high incidence rate and mortality. Abnormal regulation of gene expression plays an important role in breast cancer occurrence and development. RNA-binding proteins (RBPs) are one kind of the key regulators for gene expression. By interacting with RNA, RBPs are widely involved in RNA cutting, transport, editing, intracellular localization, and translation regulation. RBPs are important during breast cancer occurrence and progression by engaging in many aspects, like proliferation, migration, invasion, and stemness. Therefore, comprehensively understanding the role of RBPs in breast cancer progression can facilitate early diagnosis, timely treatment, and long-term survival and quality of life of breast cancer patients.
Collapse
Affiliation(s)
- Ying Chen
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Hai Qin
- Department of Clinical Laboratory, Guizhou Provincial Orthopedic Hospital, Guiyang, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
Zhang Y, Lin W, Jiang W, Wang Z. MicroRNA-18 facilitates the stemness of gastric cancer by downregulating HMGB3 though targeting Meis2. Bioengineered 2022; 13:9959-9972. [PMID: 35416122 PMCID: PMC9161930 DOI: 10.1080/21655979.2022.2062529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The recurrence and metastasis of gastric cancer are related to the stemness of gastric cancer cells. Researches have shown that miR-18 level is negatively correlated to the occurrence and development of certain cancer types. However, the effects of miR-18 on the stemness of gastric cancer remain uncertain. In this research, gastric cancer cell lines with stable overexpression of miR-18 were constructed through lentivirus infection. CCK-8 assay, RT-qPCR, Western blot, flow cytometry, and in vivo tumorigenesis assays were performed to evaluate the effects of miR-18 on the stemness of gastric cancer cells. Moreover, luciferase reporter assays found that Meis2 was the target of miR-18. Furthermore, we also found that the low-expressed oncogene HMGB3 is involved in this miR-18/Meis2 axis to further promote the stemness of gastric cancer cells. These findings suggest that the miR-18/Meis2/HMGB3 axis may be potential prognostic indicators for patients with gastric cancer.
Collapse
Affiliation(s)
- Yingjun Zhang
- Oncology Department of Radiotherapy, Zhongshan Hospital of Xiamen University, Xiamen, Fujian China
| | - Weijian Lin
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
| | - Weiping Jiang
- Oncology Department of Radiotherapy, Zhongshan Hospital of Xiamen University, Xiamen, Fujian China
| | - Zhenfa Wang
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
12
|
Liu J, Xu J, Luo B, Tang J, Hou Z, Zhu Z, Zhu L, Yao G, Li C. Immune Landscape and an RBM38-Associated Immune Prognostic Model with Laboratory Verification in Malignant Melanoma. Cancers (Basel) 2022; 14:cancers14061590. [PMID: 35326741 PMCID: PMC8946480 DOI: 10.3390/cancers14061590] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The primary treatment of malignant melanoma is a classical regimen of surgery combined with chemotherapy, targeted drugs, and immunotherapy. The purpose of this study was to explore the immune response mechanism of RNA binding protein RBM38 in the development of melanoma with the screening of effective immunodiagnostic models and targeted therapy. We found that RBM38, as an oncogene, promotes the proliferation, invasion, and migration of melanoma cells and is associated with immune infiltration and pathways. Our investigation presented the prognostic significance of RBM38-associated immune signature. In addition, this model may provide a potential strategy for improving the survival and immunotherapy of melanoma patients. Abstract Background: Current studies have revealed that RNA-binding protein RBM38 is closely related to tumor development, while its role in malignant melanoma remains unclear. Therefore, this research aimed to investigate the function of RBM38 in melanoma and the prognosis of the disease. Methods: Functional experiments (CCK-8 assay, cell colony formation, transwell cell migration/invasion experiment, wound healing assay, nude mouse tumor formation, and immunohistochemical analysis) were applied to evaluate the role of RBM38 in malignant melanoma. Immune-associated differentially expressed genes (DEGs) on RBM38 related immune pathways were comprehensively analyzed based on RNA sequencing results. Results: We found that high expression of RBM38 promoted melanoma cell proliferation, invasion, and migration, and RBM38 was associated with immune infiltration. Then, a five-gene (A2M, NAMPT, LIF, EBI3, and ERAP1) model of RBM38-associated immune DEGs was constructed and validated. Our signature showed superior prognosis capacity compared with other melanoma prognostic signatures. Moreover, the risk score of our signature was connected with the infiltration of immune cells, immune-regulatory proteins, and immunophenoscore in melanoma. Conclusions: We constructed an immune prognosis model using RBM38-related immune DEGs that may help evaluate melanoma patient prognosis and immunotherapy modalities.
Collapse
Affiliation(s)
- Jinfang Liu
- Department of Plastic and Burns Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 GuangZhou Rd, Nanjing 210029, China; (J.L.); (B.L.); (J.T.); (Z.H.); (Z.Z.)
| | - Jun Xu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Soochow 213000, China;
| | - Binlin Luo
- Department of Plastic and Burns Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 GuangZhou Rd, Nanjing 210029, China; (J.L.); (B.L.); (J.T.); (Z.H.); (Z.Z.)
| | - Jian Tang
- Department of Plastic and Burns Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 GuangZhou Rd, Nanjing 210029, China; (J.L.); (B.L.); (J.T.); (Z.H.); (Z.Z.)
| | - Zuoqiong Hou
- Department of Plastic and Burns Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 GuangZhou Rd, Nanjing 210029, China; (J.L.); (B.L.); (J.T.); (Z.H.); (Z.Z.)
| | - Zhechen Zhu
- Department of Plastic and Burns Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 GuangZhou Rd, Nanjing 210029, China; (J.L.); (B.L.); (J.T.); (Z.H.); (Z.Z.)
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China;
| | - Gang Yao
- Department of Plastic and Burns Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 GuangZhou Rd, Nanjing 210029, China; (J.L.); (B.L.); (J.T.); (Z.H.); (Z.Z.)
- Correspondence: (G.Y.); (C.L.)
| | - Chujun Li
- Department of Plastic and Burns Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 GuangZhou Rd, Nanjing 210029, China; (J.L.); (B.L.); (J.T.); (Z.H.); (Z.Z.)
- Correspondence: (G.Y.); (C.L.)
| |
Collapse
|
13
|
Zhang W, Liu L, Zhao S, Chen L, Wei Y, Chen W, Ge F. Research progress on RNA‑binding proteins in breast cancer (Review). Oncol Lett 2022; 23:121. [PMID: 35261635 PMCID: PMC8867207 DOI: 10.3892/ol.2022.13241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022] Open
Abstract
Breast cancer is the most common malignancy among women, and the abnormal regulation of gene expression serves an important role in its occurrence and development. However, the molecular mechanisms underlying gene expression are highly complex and heterogeneous, and RNA-binding proteins (RBPs) are among the key regulatory factors. RBPs bind targets in an environment-dependent or environment-independent manner to influence mRNA stability and the translation of genes involved in the formation, progression, metastasis and treatment of breast cancer. Due to the growing interest in these regulators, the present review summarizes the most influential studies concerning RBPs associated with breast cancer to elucidate the role of RBPs in breast cancer and to assess how they interact with other key pathways to provide new molecular targets for the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Wenzhu Zhang
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Linlin Liu
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Shengdi Zhao
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Liang Chen
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yuxian Wei
- Department of Endocrine Breast Surgery, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wenlin Chen
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Fei Ge
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
14
|
Qin T, Cheng Y, Wang X. RNA-binding proteins as drivers of AML and novel therapeutic targets. Leuk Lymphoma 2022; 63:1045-1057. [PMID: 35075986 DOI: 10.1080/10428194.2021.2008381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Acute myeloid leukemia (AML) is a group of genetically complex and heterogeneous invasive hematological malignancies with a low 5-year overall survival rate of 30%, which highlights the urgent need for improved treatment measures. RNA-binding proteins (RBPs) regulate the abundance of isoforms of related proteins by regulating RNA splicing, translation, stability, and localization, thereby affecting cell differentiation and self-renewal. It is increasingly believed that RBPs are essential for normal hematopoiesis, and RBPs play a key role in hematological tumors, especially AML, by acting as oncogenes or tumor suppressors. In addition, targeting an RBP that is significantly related to AML can trigger the apoptosis of leukemic stem cells or promote the proliferation of stem and progenitor cells by modulating the expression of important pathway regulatory factors such as HOXA9, MYC, and CDKN1A. Accordingly, RBPs involved in normal myeloid differentiation and the occurrence of AML may represent promising therapeutic targets.
Collapse
Affiliation(s)
- Tingyu Qin
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying Cheng
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Li X, Jia Q, Zhou Y, Jiang X, Song L, Wu Y, Wang A, Chen W, Wang S, Lu Y. Tanshinone IIA attenuates the stemness of breast cancer cells via targeting the miR-125b/STARD13 axis. Exp Hematol Oncol 2022; 11:2. [PMID: 35057866 PMCID: PMC8781032 DOI: 10.1186/s40164-022-00255-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Tanshinone II A is an effective component extracted from Salvia miltiorrhiza and the roles of Tanshinone IIA in regulating the stemness of tumor cells remain unclear. This work aims to explore the roles and underlying mechanisms of Tanshinone IIA in breast cancer stemness. METHODS In vitro mammary spheroid formation, flow cytometry assay on CD24-/CD44+ sub-population, ALDH activity detection, cell viability assay and western blot analysis, and in vivo tumor-initiating analysis were performed to examine the effects of Tanshinone IIA on the stemness of breast cancer cells. MiRNAs-based transcriptome sequencing and data analysis, online dataset analysis, luciferase reporter assay combined with rescuing experiments were constructed to explore the underlying mechanisms. RESULTS Tanshinone IIA attenuated the stemness of breast cancer cells, evident by downregulating the expression of stemness markers, hindering the capacity of spheroid formation, decreasing the CD24-/CD44+ sub-population in a concentration-dependent manner and reducing the tumor-initiating ability of breast cancer cells. Additionally, Tanshinone IIA enhanced adriamycin sensitivity and attenuated adriamycin resistance of breast cancer cells. Combined with miRNAs-based transcriptome sequencing assay, it was found that Tanshinone IIA downregulated miR-125b level and upregulated its target gene STARD13 (StAR-related lipid transfer protein 13) level, thus inactivating the miR-125b/STARD13 axis, which had been previously confirmed to promote breast cancer progression. Notably, miR-125b overexpression enhanced the stemness of breast cancer cells, and miR-125b overexpression or STARD13 knockdown impaired the inhibitory effects of Tanshinone IIA on the stemness of breast cancer cells. CONCLUSIONS Tanshinone IIA could attenuate the stemness of breast cancer cells via targeting the miR-125b/STARD13 axis.
Collapse
Affiliation(s)
- Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qi Jia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yinyin Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xuan Jiang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li Song
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuanyuan Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Shijun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China. .,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China. .,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
16
|
RBM38 is negatively regulated by miR-320b and enhances Adriamycin resistance in breast cancer cells. Oncol Lett 2021; 23:27. [PMID: 34868364 PMCID: PMC8630814 DOI: 10.3892/ol.2021.13145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is a common type of malignant tumor that is frequently accompanied by drug resistance, which is a significant challenge in the treatment of BC. Adriamycin (ADM) is a commonly used drug for the treatment of BC. The aim of the present study was to demonstrate the association between RNA binding motif protein 38 (RBM38) and ADM resistance in BC. The results revealed that the expression levels of RBM38 were significantly upregulated in ADM-resistant BC tissues and the ADM-resistant cell line, MCF-7/A, as demonstrated using reverse transcription-quantitative PCR and western blotting. In addition, the results of the MTT assay revealed that the overexpression of RBM38 enhanced the resistance of MCF-7/A cells to ADM, promoted invasiveness, as determined using a Transwell assay, inhibited the apoptosis of resistant cells, as determined using flow cytometry, and accelerated cell cycle progression from the G0 to the S phase. The results of the dual luciferase reporter assay demonstrated the binding relationship between microRNA (miR)-320b and RBM38, and the expression levels of miR-320b were significantly downregulated in ADM-resistant BC tissues and MCF-7/A cells. Overexpression of miR-320b reversed ADM resistance, suppressed invasiveness, promoted apoptosis and arrested MCF-7/A cells in the G0 phase. In addition, RBM38 was discovered to be negatively regulated by miR-320b, which was able to restore the sensitivity of BC cells to ADM by downregulating RBM38. Further exploration of the underlying regulatory mechanism revealed that the miR-320b/RBM38 signaling axis mediated the development of ADM resistance in BC by altering the expression of cell cycle-, drug resistance- and PI3K/AKT signaling pathway-related proteins. In conclusion, the results of the present study suggested that RBM38 may be negatively regulated by miR-320b, which accelerates drug resistance in BC.
Collapse
|
17
|
Ji CM, Zhang X, Fang W, Meng L, Wei X, Lu C. RNA-binding protein RNPC1 acts as an oncogene in gastric cancer by stabilizing aurora kinase B mRNA. Exp Cell Res 2021; 406:112741. [PMID: 34302858 DOI: 10.1016/j.yexcr.2021.112741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND RNPC1 is reported to act as a tumor suppressor by binding and regulating the expression of target genes in various cancers. However, the role of RNPC1 in gastric cancer and the underlying mechanisms are still unclear. METHODS Gastric cancer cells were stably transfected with lentivirus. Proliferation, migration, invasion, cell cycle in vitro and tumorigenesis in vivo were used to assess the role of RNPC1. Quantitative real-time PCR, western blotting and immunohistochemistry were used to detect the relationship between RNPC1 and aurora kinase B (AURKB). RNA immunoprecipitation (RIP), RNA electrophoretic mobility shift assays (REMSAs), and dual-luciferase reporter assays were used to identify the direct binding sites of RNPC1 with AURKB mRNA. A CCK-8 assay was conducted to confirm the function of AURKB in RNPC1-induced growth promotion. RESULTS High RNPC1 expression was found in gastric cancer tissues and cell lines and was associated with high TNM stage. RNPC1 overexpression significantly promoted the proliferation, migration, and invasion of gastric cancer cells. Knockdown of RNPC1 could impede gastric cancer tumorigenesis in nude mice. AURKB expression was positively related to RNPC1. RNPC1 directly binds to the 3'-untranslated region (3'-UTR) of AURKB and enhances AURKB mRNA stability. AURKB reversed the proliferation induced by RNPC1 in gastric cancer cells. RNPC1 resulted in mitotic defects, aneuploidy and chromosomal instability in gastric cancer cells, similar to AURKB. CONCLUSION RNPC1 acts as an oncogene in gastric cancer by influencing cell mitosis by increasing AURKB mRNA stability, which may provide a potential biomarker and a therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Chun-Mei Ji
- Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China; Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Xu Zhang
- Jiangsu Breast Disease Center, The First Affliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Wentong Fang
- Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Ling Meng
- Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Xiaolong Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China.
| | - Chen Lu
- Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
18
|
Integrated Analysis of the Roles of RNA Binding Proteins and Their Prognostic Value in Clear Cell Renal Cell Carcinoma. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:5568411. [PMID: 34306592 PMCID: PMC8263288 DOI: 10.1155/2021/5568411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/02/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022]
Abstract
Methods We downloaded the RNA sequencing data of ccRCC from the Cancer Genome Atlas (TCGA) database and identified differently expressed RBPs in different tissues. In this study, we used bioinformatics to analyze the expression and prognostic value of RBPs; then, we performed functional analysis and constructed a protein interaction network for them. We also screened out some RBPs related to the prognosis of ccRCC. Finally, based on the identified RBPs, we constructed a prognostic model that can predict patients' risk of illness and survival time. Also, the data in the HPA database were used for verification. Results In our experiment, we obtained 539 ccRCC samples and 72 normal controls. In the subsequent analysis, 87 upregulated RBPs and 38 downregulated RBPs were obtained. In addition, 9 genes related to the prognosis of patients were selected, namely, RPL36A, THOC6, RNASE2, NOVA2, TLR3, PPARGC1A, DARS, LARS2, and U2AF1L4. We further constructed a prognostic model based on these genes and plotted the ROC curve. This ROC curve performed well in judgement and evaluation. A nomogram that can judge the patient's life span is also made. Conclusion In conclusion, we have identified differentially expressed RBPs in ccRCC and carried out a series of in-depth research studies, the results of which may provide ideas for the diagnosis of ccRCC and the research of new targeted drugs.
Collapse
|
19
|
Liu H, Qin H, Zhou Y, Yuan Y, Liu Y, Chen Y, Yang Y, Ni H, Xi T, Zheng L. HET0016 attenuates the stemness of breast cancer cells through targeting CYP4Z1. Mol Carcinog 2021; 60:413-426. [PMID: 33866606 DOI: 10.1002/mc.23302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 01/06/2023]
Abstract
Ours and other previous studies have shown that CYP4Z1 is specifically and highly expressed in breast cancer, and acts as a promoter for the stemness of breast cancer cells. Here, we explored whether targeting CYP4Z1 could attenuate the stemness of breast cancer cells using HET0016, which has been confirmed to be an inhibitor of CYP4Z1 by us and others. Using the transcriptome-sequencing analysis, we found that HET0016 suppressed the expression of cancer stem cell (CSC) markers and stem cell functions. Additionally, HET0016 indeed reduced the stemness of breast cancer cells, as evident by the decrease of stemness marker expression, CD44+ /CD24- subpopulation with stemness, mammary-spheroid formation, and tumor-initiating ability. Moreover, HET0016 suppressed the metastatic capability through in vitro and in vivo experiments. Furthermore, we confirmed that HET0016 suppressed CYP4Z1 activity, and HET0016-induced inhibition on the stemness and metastasis of breast cancer cells was rescued by CYP4Z1 overexpression. Thus, our results demonstrate that HET0016 can attenuate the stemness of breast cancer cells through targeting CYP4Z1.
Collapse
Affiliation(s)
- Hai Liu
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hai Qin
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yi Zhou
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yin Yuan
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yichen Liu
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ying Chen
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yue Yang
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Haiwei Ni
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Tao Xi
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Lufeng Zheng
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
20
|
Wu B, Yang C, Fang Y, Ding W, Zhang Y. Long noncoding RNA DUXAP10 promotes the stemness of glioma cells by recruiting HuR to enhance Sox12 mRNA stability. ENVIRONMENTAL TOXICOLOGY 2021; 36:840-849. [PMID: 33340249 DOI: 10.1002/tox.23087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/09/2020] [Indexed: 06/12/2023]
Abstract
Long noncoding RNA (lncRNA) DUXAP10 has been shown to act as an oncogene in various tumors; however, its roles in glioma progression have never been established. Here, we show that DUXAP10 is overexpressed in glioma tissues and cells. Loss of function experiments reveal that DUXAP10 knockdown has little effects on glioma cell viability, but significantly reduces the stemness of glioma cells, which is characterized as the decrease of stemness marker expression, tumor sphere-forming ability, and ALDH activity. RNA immunoprecipitation and immunofluorescence assays indicate that DUXAP10 can directly interact with HuR protein and suppress the cytoplasm-nuclear translocation of HuR, which subsequently enhances Sox12 mRNA stability in cytoplasm and thus increases Sox12 expression. Further rescuing experiments show that the HuR/Sox12 axis is responsible for DUXAP10-mediated effects on glioma cell stemness.
Collapse
Affiliation(s)
- Bolin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chengyou Yang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuefeng Fang
- International College, Guangdong University of Foreign Studies, Guangzhou, China
| | - Weilong Ding
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuqing Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Wang Y, Zhou X, Han P, Lu Y, Zhong X, Yang Y, Li D, Liu D, Li Q, Pan N, Mo Y, Luo W, Li P, Zhou X, Liudmila M. Inverse correlation of miR-27a-3p and CDH5 expression serves as a diagnostic biomarker of proliferation and metastasis of clear cell renal carcinoma. Pathol Res Pract 2021; 220:153393. [PMID: 33740544 DOI: 10.1016/j.prp.2021.153393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Cadherin-5 (CDH5) is aberrantly expressed in a variety of human cancers and plays an important role in angiogenesis. The present study provides further insight into the role of miR-27a-3p in the regulation of CDH5 expression in renal clear cell carcinoma (ccRCC). METHODS Thedysregulation of CDH5 expression in ccRCC and its association with clinicopathological characteristics were analyzed using the TCGA database. A meta-analysis was performed to verify the alteration of CDH5 expression in ccRCC using the GEO database. Quantitative RT-PCR and immunohistochemical staining were applied to assess the transcriptional and protein levels of CDH5. TargetScan and Tarbase were employed to predict the miRNAs with the potential to target mRNA of CDH5. RESULTS The mRNA level of CDH5 in ccRCCwas significantly higher than in normal tissue. CDH5 mRNA expression could therefore serve as a potential diagnostic biomarker for ccRCC (AUC = 0.844). However, the reduced CDH5 transcription levels were significantly correlated with patients in the T3-4 stage, lymph node, and distant metastasis, as well as with a worse clinical outcome. We further observed that CDH5, at the protein level, was almost absent in ccRCC samples. In addition, a few databases screen showed that mir-27a-3p is a highly conserved miRNA targeting CDH5. The expression of mir-27a-3p was significantly elevated in ccRCC tissues in contrast to normal tissues. Importantly, it was positively associated with the T3-4 stage and M stage, respectively, suggesting that the expression level of mir-27a-3p could serve as a diagnostic biomarker for ccRCC (AUC = 0.775). CONCLUSION Our data suggest that thereduced translational level of CDH5 in ccRCC was related to the overexpression of mir-27a-3p. The higher mir-27a-3p and lower CDH5 expression significantly correlated with advanced clinical stages for ccRCC patients.
Collapse
Affiliation(s)
- Yifang Wang
- Life Science Institute, Guangxi Medical University, China
| | - Xiaohui Zhou
- Life Science Institute, Guangxi Medical University, China
| | - Peipei Han
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Yunliang Lu
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xuemin Zhong
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Yanping Yang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Danping Li
- Life Science Institute, Guangxi Medical University, China
| | - Deling Liu
- Life Science Institute, Guangxi Medical University, China
| | - Qiuyun Li
- Life Science Institute, Guangxi Medical University, China
| | - Nenghui Pan
- Life Science Institute, Guangxi Medical University, China
| | - Yingxi Mo
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Wenqi Luo
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Ping Li
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xiaoying Zhou
- Life Science Institute, Guangxi Medical University, China.
| | - Matskova Liudmila
- Institute of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad, Russia.
| |
Collapse
|
22
|
Jaafar L, Fakhoury I, Saab S, El-Hajjar L, Abou-Kheir W, El-Sibai M. StarD13 differentially regulates migration and invasion in prostate cancer cells. Hum Cell 2021; 34:607-623. [PMID: 33420961 DOI: 10.1007/s13577-020-00479-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/24/2020] [Indexed: 11/26/2022]
Abstract
Prostate cancer is the second most commonly diagnosed cancer in men and one of the main leading causes of cancer deaths among men worldwide. Rapid uncontrolled growth and the ability to metastasize to other sites are key hallmarks in cancer development and progression. The Rho family of GTPases and its activators the GTPase-activating proteins (GAPs) are required for regulating cancer cell proliferation and migration. StarD13 is a GAP for Rho GTPases, specifically for RhoA and Cdc42. We have previously shown that StarD13 acts as a tumor suppressor in astrocytoma as well as breast and colorectal cancer. In this study, we performed a functional comparative analysis of StarD13 targets/and or interacting molecules to understand the general role that StarD13 plays in cancers. Our data highlight the importance of StarD13 in modulating several hallmarks of cancer. Findings from database mining and immunohistochemistry revealed that StarD13 is underexpressed in prostate cancers, in addition knocking down Stard13 increased cancer cell proliferation, consistent with its role as a tumor suppressor. Stard13 depletion, however, led to an increase in cell adhesion, which inhibited 2D cell migration. Most interestingly, StarD13 depletion increases invasion and matrix degradation, at least in part, through its regulation of Cdc42. Altogether, the data presented suggest that StarD13 acts as a tumor suppressor inhibiting prostate cancer cell invasion.
Collapse
Affiliation(s)
- Leila Jaafar
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Chouran, P.O. Box 13-5053, Beirut, 1102 2801, Lebanon
| | - Isabelle Fakhoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Chouran, P.O. Box 13-5053, Beirut, 1102 2801, Lebanon
| | - Sahar Saab
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Chouran, P.O. Box 13-5053, Beirut, 1102 2801, Lebanon
| | - Layal El-Hajjar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Chouran, P.O. Box 13-5053, Beirut, 1102 2801, Lebanon.
| |
Collapse
|
23
|
Zou C, Wan Y, He L, Zheng JH, Mei Y, Shi J, Zhang M, Dong Z, Zhang D. RBM38 in cancer: role and mechanism. Cell Mol Life Sci 2021; 78:117-128. [PMID: 32642788 PMCID: PMC11072576 DOI: 10.1007/s00018-020-03593-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022]
Abstract
Cancer is the second leading cause of death globally. Abnormity in gene expression regulation characterizes the trajectory of tumor development and progression. RNA-binding proteins (RBPs) are widely dysregulated, and thus implicated, in numerous human cancers. RBPs mainly regulate gene expression post-transcriptionally, but emerging studies suggest that many RBPs can impact transcription by acting on chromatin as transcription factors (TFs) or cofactors. Here, we review the evidence that RBM38, an intensively studied RBP, frequently plays a tumor-suppressive role in multiple human cancer types. Genetic studies in mice deficient in RBM38 on different p53 status also establish RBM38 as a tumor suppressor (TS). By uncovering a spectrum of transcripts bound by RBM38, we discuss the diversity in its mechanisms of action in distinct biological contexts. Examination of the genomic features and expression pattern of RBM38 in human tissues reveals that it is generally lost but rarely mutated, in cancers. By assessing future trends in the study of RBM38 in cancer, we signify the possibility of targeting RBM38 and its related pathways as therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Cheng Zou
- College of Biology, Hunan University, Changsha, 410082, China
| | - Ying Wan
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lingjing He
- College of Biology, Hunan University, Changsha, 410082, China
| | - Jin Hai Zheng
- College of Biology, Hunan University, Changsha, 410082, China
| | - Yang Mei
- College of Biology, Hunan University, Changsha, 410082, China
| | - Junfeng Shi
- College of Biology, Hunan University, Changsha, 410082, China
| | - Min Zhang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhiqiang Dong
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Dingxiao Zhang
- College of Biology, Hunan University, Changsha, 410082, China.
| |
Collapse
|
24
|
Yao J, Lu X, Wang Y, Li J, Ni B. Long noncoding RNAs AC026904.1 is essential for TGF-β-induced migration and epithelial-mesenchymal transition through functioning as an enhancer of Slug in lung cancer cells. ENVIRONMENTAL TOXICOLOGY 2020; 35:942-951. [PMID: 32270919 DOI: 10.1002/tox.22930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/14/2020] [Accepted: 03/27/2020] [Indexed: 06/11/2023]
Abstract
Long noncoding RNA (lncRNA) AC026904.1 has been confirmed to be necessary for breast cancer metastasis. This work aims to investigate the effects of lncRNA AC026904.1 on lung cancer metastasis. We found that lncRNA AC026904.1 displayed a higher level in metastatic lung cancer tissues than adjacent tissues and nonmetastatic lung cancer tissues, and lung cancer cells treated with TGF-β. The expression of AC026904.1 was increased by the non-canonical TGF-β signaling. Additionally, AC026904.1 acts as an enhancer of the key metastatic factor Slug in the nucleus. This AC026904.1/Slug axis is necessary for TGF-β-mediated migration and epithelial-mesenchymal transition in lung cancer cells. This work firstly uncovers that AC026904.1 increases Slug expression at transcriptional level and subsequently plays critical effects in lung cancer metastasis, providing novel evidences that AC026904.1 holds great potential to be used as a marker for metastatic lung cancer.
Collapse
Affiliation(s)
- Jie Yao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaochen Lu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxuan Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiaxi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Ni
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
25
|
Yin X, Fang T, Zhang L, Lin X, Yang Y, Lou S, Li C, Yu X, Xue Y. Impact of CD144 gene expression on outcomes in stage III gastric cancer patients. Pathology 2020; 52:657-669. [PMID: 32859388 DOI: 10.1016/j.pathol.2020.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/06/2020] [Accepted: 05/13/2020] [Indexed: 10/23/2022]
Abstract
CD144 has been shown to promote tumour angiogenesis, invasion and metastasis in malignant tumours. The purpose of the present study was to investigate the clinical prognostic significance of CD144 in advanced gastric cancer (GC) to complement the American Joint Committee on Cancer (AJCC) 8th Edition convention. We established that CD144 was highly related to angiogenesis using The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) public databases. We randomly selected 173 stage III GC patients who received curative gastrectomy. The expression level of CD144 was assessed by immunohistochemistry and Image-Pro Plus software. After survival analysis, nomograms were created to predict the risk of stage III gastric cancer patients' 5-years survival. In this study, the median value of the CD144 positive area/total area under the microscope was 5.6%, and this was defined as the cut-off value. The expression of CD144 assisted further subgrouping of stage Ⅲa, Ⅲb, and Ⅲc GC patients. To evaluate the disease-free survival (DFS) and overall survival (OS) of patients, univariate and multivariate analysis was performed, which showed that the expression of CD144 was an independent predictor for DFS, and Borrmann type and expression of CD144 were independent predictors for OS (p<0.05). Nomograms were used to evaluate the risk of stage III GC by combining Borrmann type and the expression level of CD144. In advanced GC patients, the expression level of CD144 is a useful prognostic indicator in evaluating the risk of disease prognosis.
Collapse
Affiliation(s)
- Xin Yin
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Tianyi Fang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Lei Zhang
- Department of Pathology, Harbin Medical University, Harbin, P.R. China
| | - Xuan Lin
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Yongheng Yang
- Department of Pathology, Harbin Medical University, Harbin, P.R. China
| | - Shenghan Lou
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Chunfeng Li
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Xuefeng Yu
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Yingwei Xue
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China.
| |
Collapse
|
26
|
Qin H, Ni H, Liu Y, Yuan Y, Xi T, Li X, Zheng L. RNA-binding proteins in tumor progression. J Hematol Oncol 2020; 13:90. [PMID: 32653017 PMCID: PMC7353687 DOI: 10.1186/s13045-020-00927-w] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/25/2020] [Indexed: 02/08/2023] Open
Abstract
RNA-binding protein (RBP) has a highly dynamic spatiotemporal regulation process and important biological functions. They are critical to maintain the transcriptome through post-transcriptionally controlling the processing and transportation of RNA, including regulating RNA splicing, polyadenylation, mRNA stability, mRNA localization, and translation. Alteration of each process will affect the RNA life cycle, produce abnormal protein phenotypes, and thus lead to the occurrence and development of tumors. Here, we summarize RBPs involved in tumor progression and the underlying molecular mechanisms whereby they are regulated and exert their effects. This analysis is an important step towards the comprehensive characterization of post-transcriptional gene regulation involved in tumor progression.
Collapse
Affiliation(s)
- Hai Qin
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Haiwei Ni
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Yichen Liu
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Yaqin Yuan
- Guizhou Medical Device Testing Center, Guiyang, 550004, Guizhou, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
27
|
Wang Y, Wei C, Yang Y, Luo A, Zhang X, Zheng D, Lu X, Zhang K, Duan X, Xu X. Hepatocyte nuclear factor-1β suppresses the stemness and migration of colorectal cancer cells through promoting miR-200b activity. Mol Carcinog 2020; 59:989-999. [PMID: 32495507 DOI: 10.1002/mc.23229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/06/2020] [Accepted: 05/20/2020] [Indexed: 01/23/2023]
Abstract
The effects of hepatocyte nuclear factors (HNFs) have been established in various tumors; however, the roles of HNF-1β in colorectal cancer progression are never been found. In the present study, HNF-1β expression was initially detected in clinical tissue samples and online datasets and HNF-1β was found to be highly expressed in colorectal cancer tissues. In addition, a positive correlation existed between HNF-1β expression and the overall survival of patients with colorectal cancer. In vitro and in vivo experiments revealed that HNF-1β suppressed the stemness and migration of colorectal cancer cells. Combined with microRNAs (miRNAs) based on transcriptome-sequencing analysis, mechanistic studies showed that HNF-1β directly bound to miR-200b promoter and thus promoted miR-200b expression, this HNF-1β/miR-200b resulted in the downregulation of the expression of miR-200b downstream effectors. Furthermore, HNF-1β inhibits the stemness and migration of colorectal cancer cells through miR-200b. This study reveals a novel HNF-1β/miR-200b axis responsible for the stemness of colorectal cancer cells.
Collapse
Affiliation(s)
- Yuhui Wang
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Chengqiong Wei
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Yingying Yang
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Ailin Luo
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Xiyang Zhang
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Dongxuan Zheng
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Xi Lu
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Kefeng Zhang
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Xiaoqun Duan
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| | - Xiaotian Xu
- Guangxi Colleges and Universities Key Laboratory of Pharmacology, Guilin Medical University, Guilin, China
| |
Collapse
|
28
|
Wen X, Gao L, Hu Y. LAceModule: Identification of Competing Endogenous RNA Modules by Integrating Dynamic Correlation. Front Genet 2020; 11:235. [PMID: 32256525 PMCID: PMC7093494 DOI: 10.3389/fgene.2020.00235] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/27/2020] [Indexed: 12/14/2022] Open
Abstract
Competing endogenous RNAs (ceRNAs) regulate each other by competitively binding microRNAs they share. This is a vital post-transcriptional regulation mechanism and plays critical roles in physiological and pathological processes. Current computational methods for the identification of ceRNA pairs are mainly based on the correlation of the expression of ceRNA candidates and the number of shared microRNAs, without considering the sensitivity of the correlation to the expression levels of the shared microRNAs. To overcome this limitation, we introduced liquid association (LA), a dynamic correlation measure, which can evaluate the sensitivity of the correlation of ceRNAs to microRNAs, as an additional factor for the detection of ceRNAs. To this end, we firstly analyzed the effect of LA on detecting ceRNA pairs. Subsequently, we proposed an LA-based framework, termed LAceModule, to identify ceRNA modules by integrating the conventional Pearson correlation coefficient and dynamic correlation LA with multi-view non-negative matrix factorization. Using breast and liver cancer datasets, the experimental results demonstrated that LA is a useful measure in the detection of ceRNA pairs and modules. We found that the identified ceRNA modules play roles in cell adhesion, cell migration, and cell-cell communication. Furthermore, our results show that ceRNAs may represent potential drug targets and markers for the treatment and prognosis of cancer.
Collapse
Affiliation(s)
- Xiao Wen
- School of Computer Science and Technology, Xidian University, Xi'an, China
| | - Lin Gao
- School of Computer Science and Technology, Xidian University, Xi'an, China
| | - Yuxuan Hu
- School of Computer Science and Technology, Xidian University, Xi'an, China
| |
Collapse
|
29
|
Wu X, Wang Y, Zhong W, Cheng H, Tian Z. RNA Binding Protein RNPC1 Suppresses the Stemness of Human Endometrial Cancer Cells via Stabilizing MST1/2 mRNA. Med Sci Monit 2020; 26:e921389. [PMID: 32088727 PMCID: PMC7055196 DOI: 10.12659/msm.921389] [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] [Indexed: 12/29/2022] Open
Abstract
Background RNA binding protein RNPC1 has a tumor-suppressive role in various tumors, nevertheless, the role of RNPC1 in human endometrial cancer (EC) are never been reported. Material/Methods Western blot, quantitative polymerase chain reaction and sphere forming analysis were performed to evaluate the stem-like traits of cells and RNPC1-induced effects on EC cell stemness. RNA immunoprecipitation (RIP) was constructed to investigate the underlying mechanisms. Results The spheres formed by EC cells, named EC spheres, exhibited a remarkably higher stemness than the parental cells, which is characterized as the increase of sphere forming ability, ALDH1 activity, stemness marker expression and migration ability. Notably, RNPC1 expression was decreased in poorly differentiated EC cells than that in EC cells with moderately differentiated. Additionally, RNPC1 expression was significantly decreased in EC spheres and RNPC1 overexpression attenuated the stemness of EC spheres. Moreover, RNPC1 overexpression decreased the migration ability of EC spheres. Mechanistic studies showed that RNPC1 overexpression activated the Hippo pathway through directly binding to MST1/2. Inhibition of MST1/2 rescued RNPC1-mediated effects on EC sphere stemness. Conclusions Therefore, our results indicate a novel RNPC1/MST1/2 signaling responsible for EC cell stemness.
Collapse
Affiliation(s)
- XingMei Wu
- Department of Gynecology, The People's Hospital of Lishui, Lishui, Zhejiang, China (mainland)
| | - YongHui Wang
- Department of Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| | - WeiJuan Zhong
- Department of Gynecology, The People's Hospital of Lishui, Lishui, Zhejiang, China (mainland)
| | - HuiFei Cheng
- Department of Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| | - ZhiFeng Tian
- Department of Radiation Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| |
Collapse
|
30
|
Wu VT, Kiriazov B, Koch KE, Gu VW, Beck AC, Borcherding N, Li T, Addo P, Wehrspan ZJ, Zhang W, Braun TA, Brown BJ, Band V, Band H, Kulak MV, Weigel RJ. A TFAP2C Gene Signature Is Predictive of Outcome in HER2-Positive Breast Cancer. Mol Cancer Res 2019; 18:46-56. [PMID: 31619506 DOI: 10.1158/1541-7786.mcr-19-0359] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/05/2019] [Accepted: 10/11/2019] [Indexed: 11/16/2022]
Abstract
The AP-2γ transcription factor, encoded by the TFAP2C gene, regulates the expression of estrogen receptor-alpha (ERα) and other genes associated with hormone response in luminal breast cancer. Little is known about the role of AP-2γ in other breast cancer subtypes. A subset of HER2+ breast cancers with amplification of the TFAP2C gene locus becomes addicted to AP-2γ. Herein, we sought to define AP-2γ gene targets in HER2+ breast cancer and identify genes accounting for physiologic effects of growth and invasiveness regulated by AP-2γ. Comparing HER2+ cell lines that demonstrated differential response to growth and invasiveness with knockdown of TFAP2C, we identified a set of 68 differentially expressed target genes. CDH5 and CDKN1A were among the genes differentially regulated by AP-2γ and that contributed to growth and invasiveness. Pathway analysis implicated the MAPK13/p38δ and retinoic acid regulatory nodes, which were confirmed to display divergent responses in different HER2+ cancer lines. To confirm the clinical relevance of the genes identified, the AP-2γ gene signature was found to be highly predictive of outcome in patients with HER2+ breast cancer. We conclude that AP-2γ regulates a set of genes in HER2+ breast cancer that drive cancer growth and invasiveness. The AP-2γ gene signature predicts outcome of patients with HER2+ breast cancer and pathway analysis predicts that subsets of patients will respond to drugs that target the MAPK or retinoic acid pathways. IMPLICATIONS: A set of genes regulated by AP-2γ in HER2+ breast cancer that drive proliferation and invasion were identified and provided a gene signature that is predictive of outcome in HER2+ breast cancer.
Collapse
Affiliation(s)
- Vincent T Wu
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | - Boris Kiriazov
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | - Kelsey E Koch
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | - Vivian W Gu
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Anna C Beck
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | | | - Tiandao Li
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | - Peter Addo
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | | | - Weizhou Zhang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Terry A Braun
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa
| | - Bartley J Brown
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Hamid Band
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | | | - Ronald J Weigel
- Department of Surgery, University of Iowa, Iowa City, Iowa. .,Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa.,Department of Biochemistry, University of Iowa, Iowa City, Iowa
| |
Collapse
|
31
|
Unearthing Regulatory Axes of Breast Cancer circRNAs Networks to Find Novel Targets and Fathom Pivotal Mechanisms. Interdiscip Sci 2019; 11:711-722. [PMID: 31187432 DOI: 10.1007/s12539-019-00339-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 01/20/2023]
Abstract
Circular RNAs (circRNAs) possess valuable characteristics for both diagnosis and treatment of several human cancers including breast cancer (BC). In this study, we combined several systems, biology tools and approaches to identify influential BC circRNAs, miRNAs, and related mRNAs as the members of competing endogenous RNAs (ceRNAs) networks and related RNA binding proteins (RBPs) to study and decipher the BC-triggering biological processes and pathways. Rooting from the identified total of 25 co-differentially expressed circRNAs (DECs) between triple negative (TN) and luminal A subtypes of BC from microarray analysis, five hub DECs (hsa_circ_0003227, hsa_circ_0001955, hsa_circ_0020080, hsa_circ_0001666, and hsa_circ_0065173) and top eleven RBPs (AGO1, AGO2, EIF4A3, FMRP, HuR (ELAVL1), IGF2BP1, IGF2BP2, IGF2BP3, EWSR1, FUS, and PTB) were explored to form the upper stream regulatory elements. All the hub circRNAs were regarded as a super sponge having multiple miRNA response elements (MREs). Then, three BC leading miRNAs (hsa-miR-149, hsa-miR-182, and hsa-miR-383) were also introduced from merging several established ceRNAs networks. The predicted 7- and 8-mer MREs matches between hub circRNAs and leading miRNAs ensured their enduring regulatory capability. The mined downstream mRNAs of the circRNAs-miRNAs network then were presented to STRING database to form the PPI network and to decipher the issue from another point of view. The BC interconnected enriched pathways and processes guarantee the merits of the ceRNAs network's members as targetable therapeutic elements. This study suggested extensive panels of novel therapeutic targets that are in charge of BC progression, hence their impressive role cannot be excluded and needs deeper empirical laboratory designs.
Collapse
|
32
|
Li X, Zheng L, Xi T. WITHDRAWN: Tanshinone IIA-mediated inhibition on miR-125b/STARD13 axis attenuates the stemness and enhances adriamycin sensitivity of breast cancer cells. Biochimie 2019:S0300-9084(19)30147-6. [PMID: 31078586 DOI: 10.1016/j.biochi.2019.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 05/06/2019] [Indexed: 11/19/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Tao Xi
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China
| |
Collapse
|
33
|
Zhang L, Chen Y, Li C, Liu J, Ren H, Li L, Zheng X, Wang H, Han Z. RNA binding protein PUM2 promotes the stemness of breast cancer cells via competitively binding to neuropilin-1 (NRP-1) mRNA with miR-376a. Biomed Pharmacother 2019; 114:108772. [PMID: 30909144 DOI: 10.1016/j.biopha.2019.108772] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 12/20/2022] Open
Abstract
Others and ours studies have established the promoting roles of NRP-1 (neuropilin-1) in breast cancer, however, the underlying mechanisms by which NRP-1 is regulated are still confused. Here, bioinformatics analysis indicated that RNA binding protein PUM2 could bind to NRP-1 mRNA. Clinical samples showed that PUM2 expression was significantly increased in breast cancer tissues, negatively correlated with the overall survival and relapse-free survival of breast cancer patients, and positively correlated with NRP-1 expression. Meanwhile, PUM2 expression was remarkably increased in non-adherent spheroids. in vitro experiments demonstrated that PUM2 knockdown attenuated the stemness of breast cancer cells, evident by the decrease of spheroid formation capacity, ALDH1 activity and stemness marker expression. Mechanistically, RNA immunoprecipitation (RIP) and luciferase reporter analysis indicated that PUM2 competitively bound to NRP 3'UTR with miR-376a, which had been previously confirmed by us to suppress the stemness of breast cancer cells, and increased NRP-1 mRNA stability and expression. Furthermore, ectopic expression of NRP-1 or miR-376a knockdown rescued the inhibitory effects of NRP-1 knockdown on the stemness of breast cancer cells. Thus, our results suggest that PUM2 could facilitate the stemness of breast cancer cells by competitively binding to NRP-1 3'UTR with miR-376a.
Collapse
Affiliation(s)
- Lansheng Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Yanwei Chen
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Caihong Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Jinyang Liu
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Huiwen Ren
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Lishan Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Xia Zheng
- Department of Radiation Oncology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Hui Wang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Zhengxiang Han
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.
| |
Collapse
|
34
|
Wu H, He Y, Chen H, Liu Y, Wei B, Chen G, Lin H, Lin H. LncRNA THOR increases osteosarcoma cell stemness and migration by enhancing SOX9 mRNA stability. FEBS Open Bio 2019; 9:781-790. [PMID: 30984551 PMCID: PMC6443997 DOI: 10.1002/2211-5463.12620] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/11/2019] [Accepted: 02/20/2019] [Indexed: 12/31/2022] Open
Abstract
Although the long non-coding RNA THOR has been reported to promote cancer stem cell expansion in liver cancer and gastric cancer, its effects on osteosarcoma (OS) cells remain unclear. Here, we investigated the roles of THOR in the stemness and migration of OS cells. We report that the level of THOR is remarkably upregulated in OS cell spheroids compared to that in OS adherent cells. THOR overexpression increased spheroid formation ability and aldehyde dehydrogenase 1 (ALDH1) activity in OS adherent cells, and the opposite effect was observed in spheroids with THOR knockdown. Additionally, the spheroids formed by OS adherent cells exhibited a stronger migration ability, which was attenuated by THOR knockdown, and THOR overexpression increased OS cell migration. Mechanistically, mRNA stability, luciferase reporter, and RNA-RNA in vitro interaction assays indicated that THOR can directly bind to the middle region of the SOX9 3'-untranslated region (UTR), and enhances its mRNA stability, thereby increasing its expression. Notably, SOX9 knockdown reduced the ability of THOR overexpression to promote the stemness of OS cells. These findings indicate that the lncRNA THOR can promote the stemness and migration of OS cells by directly binding to the middle region of SOX9 3'UTR, thereby enhancing SOX9 mRNA stability and increasing its expression; thus, we provide information that may be of use in identifying potential targets for OS treatment.
Collapse
Affiliation(s)
- Haojun Wu
- The Orthopaedic Center The Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Yanxia He
- The Operating Room The Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Hang Chen
- The Orthopaedic Center The Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Yanzhi Liu
- Department of Pharmacology The Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Bo Wei
- The Orthopaedic Center The Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Guanghua Chen
- The Orthopaedic Center The Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Han Lin
- The Orthopaedic Center The Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Hao Lin
- The Orthopaedic Center The Affiliated Hospital of Guangdong Medical University Zhanjiang China
| |
Collapse
|
35
|
Gao L, Guo Q, Li X, Yang X, Ni H, Wang T, Zhao Q, Liu H, Xing Y, Xi T, Zheng L. MiR-873/PD-L1 axis regulates the stemness of breast cancer cells. EBioMedicine 2019; 41:395-407. [PMID: 30803931 PMCID: PMC6444076 DOI: 10.1016/j.ebiom.2019.02.034] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Breast cancer stem cells have self-renewal capability and are resistant to conventional chemotherapy. PD-L1 could promote the expression of stemness markers (OCT4 and Nanog) in breast cancer stem cells. However, the mechanisms by which PD-L1 regulates the stemness of breast cancer cells and PD-L1 is regulated in breast cancer cells are still unclear. METHODS Lentivirus infection was used to construct stable cell lines. The correlation between PD-L1 and stemness markers expression was evaluated in clinical samples. Additionally, luciferase reporter assay combined with RNA-Fluorescence in situ hybridization (RNA-FISH) and RNA-binding protein immunoprecipitation (RIP) assays were used to verify the direct binding of miR-873 on PD-L1. Furthermore, flow cytometry, mammosphere formation combined with nude mouse tumor xenograft model were carried out to examine the effects of miR-873/PD-L1 axis on the stemness of breast cancer cells. Finally, MTT assay was performed to determine the effects of miR-873/PD-L1 axis on drug resistance. FINDINGS PD-L1 expression was positively correlated with the expression of stemness markers, and overexpression of PD-L1 contributed to chemoresistance and stemness-like properties in breast cancer cells via activating PI3K/Akt and ERK1/2 pathways. Mechanistically, miR-873 inhibited PD-L1 expression through directly binding to its 3'-untranslated region (UTR), and miR-873 attenuated the stemness and chemoresistance of breast cancer cells which was dependent on PD-L1 and the downstream PI3K/Akt and ERK1/2 signaling. Notably, the promotion of PD-L1 on the stemness and chemoresistance was enhanced by recombinant PD-1 (rPD-1), this effect was attenuated by PD-1/PD-L1 inhibitor. INTERPRETATION miR-873/PD-L1 regulatory axis might serve as a therapeutic target to enhance the chemo-sensitivity and eliminate the stemness of breast cancer cells. FUND: This work was supported by the National Nature Science Foundation of China, No. 81702957, China Postdoctoral Science Foundation, No. 2017M620230, the Postdoctoral Research Funding Scheme of Jiangsu Province (2017), No. 1701197B, and the Priority Academic Program Development (PAPD) of Jiangsu Higher Education Institutions.
Collapse
Affiliation(s)
- Lanlan Gao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Qianqian Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Xuan Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Haiwei Ni
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Ting Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Qiong Zhao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Hai Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
36
|
SUMO protease SENP1 acts as a ceRNA for TGFBR2 and thus activates TGFBR2/Smad signaling responsible for LPS-induced sepsis. Biomed Pharmacother 2019; 112:108620. [PMID: 30797150 DOI: 10.1016/j.biopha.2019.108620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 12/11/2022] Open
Abstract
This study aims to explore the roles and related mechanisms of SUMO protease SENP1 in sepsis. Here, RNA-sequencing assay showed that SENP1 was significantly increased in human umbilical vein endothelial cells (HUVECs) with LPS treatment. Gene set enrichment analysis (GSEA) of RNA-sequencing dataset revealed that a positive enrichment of inflammation signatures was observed in HUVECs with SENP1 3'UTR overexpression. Further functional annotation analysis revealed that SENP1 3'UTR overexpression was positively correlated with TGFBR2 signaling pathway. Mechanistically, TGFBR2 was identified as a ceRNA (competing endogenous RNA) target of SENP1 and the downstream effectors Smad2/3 were also overexpressed in HUVECs with SENP1 3'UTR overexpression. Injection of SENP1 siRNA following LPS treatment attenuated LPS-induced sepsis, evidenced by the downregulation of IL-2 and TNF-α secretion and prolonged the overall survival of septic mice. Consistent results were obtained in vitro. Additionally, TGFBR2 overexpression partially abrogated SENP1 siRNA-mediated inhibition on LPS-induced sepsis. Thus, these results suggest that SENP1 promotes sepsis via activating the TGFBR2 signaling.
Collapse
|
37
|
An eight-lncRNA signature predicts survival of breast cancer patients: a comprehensive study based on weighted gene co-expression network analysis and competing endogenous RNA network. Breast Cancer Res Treat 2019; 175:59-75. [PMID: 30715658 DOI: 10.1007/s10549-019-05147-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/22/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE To identify a lncRNA signature to predict survival of breast cancer (BRCA) patients. METHODS A total of 1222 BRCA case and control datasets were downloaded from the TCGA database. The weighted gene co-expression network analysis of differentially expressed mRNAs was performed to generate the modules associated with BRCA overall survival status and further construct a hub on competing endogenous RNA (ceRNA) network. LncRNA signatures for predicting survival of BRCA patients were generated using univariate survival analyses and a multivariate Cox hazard model analysis and validated and characterized for prognostic performance measured using receiver operating characteristic (ROC) curves. RESULTS A prognostic score model of eight lncRNAs signature was identified as Prognostic score = (0.121 × EXPAC007731.1) + (0.108 × EXPAL513123.1) + (0.105 × EXPC10orf126) + (0.065 × EXPWT1-AS) + (- 0.126 × EXPADAMTS9-AS1) + (- 0.130 × EXPSRGAP3-AS2) + (0.116 × EXPTLR8-AS1) + (0.060 × EXPHOTAIR) with median score 1.088. Higher scores predicted higher risk. The lncRNAs signature was an independent prognostic factor associated with overall survival. The area under the ROC curves (AUC) of the signature was 0.979, 0.844, 0.99 and 0.997 by logistic regression, support vector machine, decision tree and random forest models, respectively, and the AUCs in predicting 1- to 10-year survival were between 0.656 and 0.748 in the test dataset from TCGA database. CONCLUSIONS The eight-lncRNA signature could serve as an independent biomarker for prediction of overall survival of BRCA. The lncRNA-miRNA-mRNA ceRNA network is a good tool to identify lncRNAs that is correlated with overall survival of BRCA.
Collapse
|