1
|
Jiang C, Luo J, Jiang X, Lv Y, Dou J. Predictive model of gene expression regulating invasion and migration of M2 macrophages in breast cancer: clinical prognosis and therapeutic implications. Transl Cancer Res 2024; 13:4187-4204. [PMID: 39262492 PMCID: PMC11384920 DOI: 10.21037/tcr-24-29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/30/2024] [Indexed: 09/13/2024]
Abstract
Background Breast cancer (BRCA) has surpassed lung cancer to become the malignant tumor with the highest incidence in female population. It occurs in malignant cells in breast tissue and is common worldwide. An increasing body of research indicates that M2 macrophages are critical to the occurrence and progression of BRCA. The aim of this work is to build a predictive model of genes related to invasion and migration of M2 macrophages, forecast the prognosis of patients with BRCA, and then evaluate the efficacy of some targeted treatments. Methods The Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/) database supplied the GSE20685 dataset, whereas the expression profile a clinical details of BRCA patients were obtained from The Cancer Genome Atlas (TCGA; https://portal.gdc.cancer.gov/) database. The genes linked to M2 macrophages and the differentially elevated genes of invasion and migration were found in GSE20685. To explore the prognosis-related invasion and migration M2 macrophage genes, the TCGA-BRCA dataset was merged with Cox regression and least absolute shrinkage and selection operator (LASSO) regression. GSE58812 was utilized for external validation. After calculating each patient's risk score, the prognostic model was examined by analyses of immune infiltration, medication sensitivity, mutation, and enrichment of the risk score. Results The risk score had a strong correlation with both several immune cells and popular anti-tumor medications. Additionally, it was discovered that the risk score was a separate prognostic factor for BRCA. Conclusions Based on invasion and migration-related M2 macrophage genes, we investigated and validated predictive characteristics in our study that may offer helpful insights into the progression and prognosis of BRCA.
Collapse
Affiliation(s)
- Chengjie Jiang
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, China
| | - Jinlei Luo
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, China
| | - Xiaoxue Jiang
- School of Chinese Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yujie Lv
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, China
| | - Jianwei Dou
- School of Pharmacy, Xi'an Jiao Tong University, Xi'an, China
| |
Collapse
|
2
|
Tang W, Gao Y, Hong S, Wang S. GFPT1 accelerates immune escape in breast cancer by modifying PD-L1 via O-glycosylation. BMC Cancer 2024; 24:1071. [PMID: 39210323 PMCID: PMC11363670 DOI: 10.1186/s12885-024-12811-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Immune escape is one of the causes of poor prognosis in breast cancer (BC). Glutamine-fructose-6-phosphate transaminase 1 (GFPT1) is the first speed-limiting enzyme of the hexosamine biosynthesis pathway (HBP) and is essential for the progression of BC. Nevertheless, the mechanism of the influence of GFPT1 in BC immune escape is not clear. METHODS First, the level of GFPT1 in BC was analyzed by starbase, and GFPT1 expression in BC tissues was measured by qRT-PCR, western blot and IHC. Then, the O-GlcNAc levels were detected by western blot. Thereafter, Co-IP was applied to examine the relationship between GFPT1 and PD-L1. At last, a mouse model was constructed for validation in vivo. RESULTS Firstly, we discovered that GFPT1 was obviously strengthened in BC. Knockdown or introduction of GFPT1 correspondingly degraded and elevated O-GlcNAc levels in cells. Further researches revealed that there was a reciprocal relationship between GFPT1 and PD-L1. Mechanistically, we disclosed that GFPT1 enhanced PD-L1 protein stability through O-glycosylation. More interestingly, GFPT1 accelerated BC cell immune escape via upregulation of O-glycosylation-modified PD-L1. In vivo, silencing of GFPT1 attenuated immune escape of BC cells by reducing PD-L1 levels. CONCLUSION GFPT1 promoted BC progression and immune escape via O-glycosylation-modified PD-L1. GFPT1 may be a potential target for BC therapy.
Collapse
Affiliation(s)
- Weifang Tang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 107, East Huanhu Road, Hefei, Anhui, 230001, China
| | - Yuan Gao
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 107, East Huanhu Road, Hefei, Anhui, 230001, China
| | - Shikai Hong
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 107, East Huanhu Road, Hefei, Anhui, 230001, China
| | - Shengying Wang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 107, East Huanhu Road, Hefei, Anhui, 230001, China.
| |
Collapse
|
3
|
Hazari V, Samali SA, Izadpanahi P, Mollaei H, Sadri F, Rezaei Z. MicroRNA-98: the multifaceted regulator in human cancer progression and therapy. Cancer Cell Int 2024; 24:209. [PMID: 38872210 DOI: 10.1186/s12935-024-03386-2] [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: 12/24/2023] [Accepted: 05/25/2024] [Indexed: 06/15/2024] Open
Abstract
MicroRNA-98 (miR-98) stands as an important molecule in the intricate landscape of oncology. As a subset of microRNAs, these small non-coding RNAs have accompanied a new era in cancer research, underpinning their significant roles in tumorigenesis, metastasis, and therapeutic interventions. This review provides a comprehensive insight into the biogenesis, molecular properties, and physiological undertakings of miR-98, highlighting its double-edged role in cancer progression-acting both as a tumor promoter and suppressor. Intriguingly, miR-98 has profound implications for various aspects of cancer progression, modulating key cellular functions, including proliferation, apoptosis, and the cell cycle. Given its expression patterns, the potential of miR-98 as a diagnostic and prognostic biomarker, especially in liquid biopsies and tumor tissues, is explored, emphasizing the hurdles in translating these findings clinically. The review concludes by evaluating therapeutic avenues to modulate miR-98 expression, addressing the challenges in therapy resistance, and assessing the efficacy of miR-98 interventions. In conclusion, while miR-98's involvement in cancer showcases promising diagnostic and therapeutic avenues, future research should pivot towards understanding its role in tumor-stroma interactions, immune modulation, and metabolic regulation, thereby unlocking novel strategies for cancer management.
Collapse
Affiliation(s)
- Vajihe Hazari
- Department of Obstetrics and Gynecology, School of Medicine, Rooyesh Infertility Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Sahar Ahmad Samali
- Department of Microbiology, Yasooj Branch, Islamic Azad University, Yasooj, Iran
| | | | - Homa Mollaei
- Department of Biology, Faculty of Sciences, University of Birjand, Birjand, Iran
| | - Farzad Sadri
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran.
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Zohreh Rezaei
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran.
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
4
|
Sun YQ, Wang B, Zheng LW, Zhao JH, Ren JG. Oral cancer cell to endothelial cell communication via exosomal miR-21/RMND5A pathway. BMC Oral Health 2024; 24:82. [PMID: 38229133 DOI: 10.1186/s12903-024-03852-3] [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: 05/04/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
Required for meiotic nuclear division 5 homolog A (RMND5A), a novel ubiquitin E3 Ligase, has been reported to correlate with poor prognosis of several cancers. However, its role in endothelial cells has not been reported. In this study, overexpression of RMND5A in human umbilical vein endothelial cells (HUVECs) was performed via lentiviral infection, followed by MTT, would healing and tube formation assay as well as signaling analysis. Moreover, crosstalk between HUVECs and oral squamous cell carcinoma (OSCC) cells was investigated by indirect co-culture with condition medium or tumor cell derived exosomes. Our results showed that overexpression of RMND5A reduced the proliferation, migration and tube formation ability of HUVECs by inhibiting the activation of ERK and NF-κB pathway. Interestingly, OSCC cells can inhibit RMND5A expression of endothelial cells via exosomal miR-21. In summary, our present study unveils that OSCC cells can activate endothelial cells via exosomal miR-21/RMND5A pathway to promote angiogenesis, which may provide novel therapeutic targets for the treatment of OSCC.
Collapse
Affiliation(s)
- Yu-Qi Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, No. 237 Luoyu Road, Wuhan, 430079, China
| | - Bing Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, No. 237 Luoyu Road, Wuhan, 430079, China
| | - Lin-Wei Zheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, No. 237 Luoyu Road, Wuhan, 430079, China
| | - Ji-Hong Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, No. 237 Luoyu Road, Wuhan, 430079, China.
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Jian-Gang Ren
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, No. 237 Luoyu Road, Wuhan, 430079, China.
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
5
|
Ren S, Yu H. The prognostic and biological importance of chromatin regulation-related genes for lung cancer is examined using bioinformatics and experimentally confirmed. Pathol Res Pract 2023; 248:154638. [PMID: 37379709 DOI: 10.1016/j.prp.2023.154638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND The pathogenesis and clinical diagnosis of lung adenocarcinoma (LUAD), a malignant illness with substantial morbidity and mortality, are still being investigated. Genes involved in chromatin regulation are crucial in the biological function of LUAD. METHODS The prognostic prediction model for LUAD was developed using multivariables and least absolute shrinkage and selection operator (LASSO) regression. It consisted of 10 chromatin regulators. The LUAD has been divided into two groups, high- and low-risk, using a predictive model. The model was shown to be accurate in predicting survival by the nomogram, receiver operating characteristic (ROC) curves, and principal component analysis (PCA). An analysis of differences in immune-cell infiltration, immunologicalfunction, and clinical traits between low- and high-risk populations was conducted. Protein-protein interaction (PPI) networks and Gene Ontology (GO) pathways of differentially expressed genes (DEGs) in the high versus low risk group were also examined to investigate the association between genes and biological pathways. The biological roles of chromatin regulators (CRs) in LUAD were finally estimated using colony formation and cell movement. The important genes' mRNA expression has been measured using real-time polymerase chain reaction (RT-PCR). RESULTS AND CONCLUSION Risk score and stage based on the model could be seen as separate prognostic indicators for patients with LUAD. The main signaling pathway difference across various risk groups was in cell cycle. The immunoinfiltration profile of the tumor microenvironment (TME) and individuals with different risk levels were correlated, suggesting that the interaction of immune cells with the tumor led to the creation of a favorable immunosuppressive microenvironment. These discoveries aid in the creation of individualized therapies for LUAD patients.
Collapse
Affiliation(s)
- Shanshan Ren
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Haiyang Yu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Deng X, Ma J, Zhou W, Yuan Y, Wang B, Meng X. GID2 Interacts With CDKN3 and Regulates Pancreatic Cancer Growth and Apoptosis. J Transl Med 2023; 103:100122. [PMID: 36828188 DOI: 10.1016/j.labinv.2023.100122] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Dysregulation of deubiquitinase or ubiquitinase-mediated protein expression contributes to various diseases, including cancer. In the present study, we identified GID2, a subunit of the glucose-induced degradation-deficient (GID) complex that functions as an E3 ubiquitin ligase, as a potential key candidate gene in pancreatic cancer (PC) progression. The functional role and potential mechanism of GID2 in PC progression were investigated. Integrated bioinformatics analysis was performed to identify differentially expressed genes in PC based on the Gene Expression Profiling Interactive Analysis data sets. We found that GID2 was upregulated in PC tissues and that a high level of GID2 expression in clinical PC samples was positively associated with tumor stage and poor survival. Functional assays elucidated that GID2 expression promoted cell growth in vitro and accelerated tumor growth in vivo. GID2 knockdown effectively attenuated the malignant behaviors of PC cells and tumor formation. Furthermore, the protein network that interacted with the GID2 protein was constructed based on the GeneMANIA website. Cyclin-dependent kinase inhibitor 3 (CDKN3), a cell cycle regulator, was identified as a potential target of the GID2 protein. We revealed that GID2 positively regulated CDKN3 expression and inhibited CDKN3 ubiquitination. Furthermore, CDKN3 downregulation reversed the promoting effects of GID2 on PC progression. Therefore, the present study demonstrated that GID2 might regulate PC progression by maintaining the stability of the CDKN3 protein. These findings highlight the potential roles of the GID2/CDKN3 axis as a potential therapeutic target in PC.
Collapse
Affiliation(s)
- Xin Deng
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jia Ma
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wenyang Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yifeng Yuan
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Baosheng Wang
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiangpeng Meng
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
7
|
Hosseini SF, Javanshir-Giv S, Soleimani H, Mollaei H, Sadri F, Rezaei Z. The importance of hsa-miR-28 in human malignancies. Biomed Pharmacother 2023; 161:114453. [PMID: 36868012 DOI: 10.1016/j.biopha.2023.114453] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
MicroRNA production in tumorigenesis is dysregulated by a variety of processes, such as proliferation and removal of microRNA genes, aberrant transcriptional regulation of microRNAs, disrupted epigenetic alterations, and failures in the miRNA biogenesis machinery. Under some circumstances, miRNAs may act as tumorigenic and maybe anti-oncogenes. Tumor aspects such as maintaining proliferating signals, bypassing development suppressors, delaying apoptosis, stimulating metastasis and invasion, and promoting angiogenesis have been linked to dysfunctional and dysregulated miRNAs. MiRNAs have been found as possible biomarkers for human cancer in a great deal of research, which requires additional evaluation and confirmation. It is known that hsa-miR-28 can function as an oncogene or tumor suppressor in many malignancies, and it does this by modulating the expression of several genes and the downstream signaling network. MiR-28-5p and miR-28-3p, which originate from the same RNA hairpin precursor miR-28, have essential roles in a variety of cancers. This review outlines the function and mechanisms of miR-28-3p and miR-28-5p in human cancers and illustrates the miR-28 family's potential utility as a diagnostic biomarker for prognosis and early detection of cancers.
Collapse
Affiliation(s)
- Seyede Fatemeh Hosseini
- Faculty Member, Tabas School of Nursing, Birjand University of Medical Sciences, Birjand, Iran
| | - Setareh Javanshir-Giv
- Faculty of Medicine, Department of Biochemistry, Birjand University of Medical Sciences, Birjand, Iran; Department of Clinical Biochemistry, Afzalipour School of Medicine & Physiology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hanieh Soleimani
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran
| | - Homa Mollaei
- Department of Biology, Faculty of Sciences, University of Birjand, Birjand, Iran
| | - Farzad Sadri
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran.
| | - Zohreh Rezaei
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran; Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
8
|
Liu Y, Han J, Kong T, Xiao N, Mei Q, Liu J. DriverMP enables improved identification of cancer driver genes. Gigascience 2022; 12:giad106. [PMID: 38091511 PMCID: PMC10716827 DOI: 10.1093/gigascience/giad106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/30/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Cancer is widely regarded as a complex disease primarily driven by genetic mutations. A critical concern and significant obstacle lies in discerning driver genes amid an extensive array of passenger genes. FINDINGS We present a new method termed DriverMP for effectively prioritizing altered genes on a cancer-type level by considering mutated gene pairs. It is designed to first apply nonsilent somatic mutation data, protein‒protein interaction network data, and differential gene expression data to prioritize mutated gene pairs, and then individual mutated genes are prioritized based on prioritized mutated gene pairs. Application of this method in 10 cancer datasets from The Cancer Genome Atlas demonstrated its great improvements over all the compared state-of-the-art methods in identifying known driver genes. Then, a comprehensive analysis demonstrated the reliability of the novel driver genes that are strongly supported by clinical experiments, disease enrichment, or biological pathway analysis. CONCLUSIONS The new method, DriverMP, which is able to identify driver genes by effectively integrating the advantages of multiple kinds of cancer data, is available at https://github.com/LiuYangyangSDU/DriverMP. In addition, we have developed a novel driver gene database for 10 cancer types and an online service that can be freely accessed without registration for users. The DriverMP method, the database of novel drivers, and the user-friendly online server are expected to contribute to new diagnostic and therapeutic opportunities for cancers.
Collapse
Affiliation(s)
- Yangyang Liu
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Jiyun Han
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Tongxin Kong
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Nannan Xiao
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Qinglin Mei
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Juntao Liu
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| |
Collapse
|
9
|
Zhang XX, Luo JH, Wu LQ. FN1 overexpression is correlated with unfavorable prognosis and immune infiltrates in breast cancer. Front Genet 2022; 13:913659. [PMID: 36035176 PMCID: PMC9417469 DOI: 10.3389/fgene.2022.913659] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/11/2022] [Indexed: 12/14/2022] Open
Abstract
Objective: To investigate the correlation of fibronectin 1 (FN1) expression with prognosis and tumor-infiltrating immune cells in breast cancer (BRCA). Methods: FN1 mRNA and protein expressions were analyzed through Tumor Immune Estimation Resource (TIMER), Gene Set Cancer Analysis (GSCA), Human Protein Atlas (HPA) databases, and immunohistochemical analysis. The clinicopathological characteristics and genetic factors affecting the FN1 mRNA expression were assessed by various public databases. Then, we analyzed the prognostic value of FN1 in BRCA by Kaplan-Meier plotter, receiver operating characteristic, and Cox regression analyses. Further, the UCSC Xena database was used to retrieve TCGA-BRCA expression profiles for functional enrichment analysis and immune cell infiltration analysis. The potential drugs for the BRCA patients with high- FN1 expression were identified using the connectivity map analysis. Results: FN1 was upregulated in BRCA tissues compared with normal tissues. High FN1 mRNA expression was correlated with poor clinical outcomes and had good performance in predicting the survival status of BRCA patients. Further, Cox regression analysis showed that FN1 was an independent prognostic factor for predicting the overall survival of patients with BRCA. Moreover, hypermethylation of FN1 contributed to a better prognosis for BRCA patients. Functional enrichment analyses revealed the ECM-receptor interaction pathway and focal adhesion as the common pathways. Moreover, FN1 showed a significant association with tumor-infiltrating immune cells and immune checkpoint inhibitors. Several drugs such as telmisartan, malotilate, and seocalcitol may have therapeutic effects in BRCA patients with high FN1 expression. Conclusion: FN1 might serve as a novel prognostic biomarker and a novel therapeutic target for BRCA. Besides, the association of FN1 with immune cells and immune checkpoint inhibitors may provide assistance for BRCA treatment.
Collapse
Affiliation(s)
- Xiu-Xia Zhang
- Department of Thyroid and Breast Surgery, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun-Hua Luo
- Department of Thyroid and Breast Surgery, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li-Qiang Wu
- Department of Hematology, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, Zhejiang, China
- *Correspondence: Li-Qiang Wu,
| |
Collapse
|
10
|
Zhang M, Xiong F, Zhang S, Guo W, He Y. Crucial Roles of miR-625 in Human Cancer. Front Med (Lausanne) 2022; 9:845094. [PMID: 35308517 PMCID: PMC8931282 DOI: 10.3389/fmed.2022.845094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/09/2022] [Indexed: 12/15/2022] Open
Abstract
Genetic and epigenetic characteristics are core factors of cancer. MicroRNAs (miRNAs) are small non-coding RNAs which regulate gene expression at the post-transcriptional level via binding to corresponding mRNAs. Recently, increasing evidence has proven that miRNAs regulate the occurrence and development of human cancer. Here, we mainly review the abnormal expression of miR-625 in a variety of cancers. In summarizing the role and potential molecular mechanisms of miR-625 in various tumors in detail, we reveal that miR-625 is involved in a variety of biological processes, such as cell proliferation, invasion, migration, apoptosis, cell cycle regulation, and drug resistance. In addition, we discuss the lncRNA-miRNA-mRNA and circRNA-miRNA-mRNA networks and briefly explain the specific mechanisms of competing endogenous RNAs. In conclusion, we reveal the potential value of miR-625 in cancer diagnosis, treatment, and prognosis and hope to provide new ideas for the clinical application of miR-625.
Collapse
Affiliation(s)
- Menggang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Fei Xiong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Yuting He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| |
Collapse
|
11
|
Chang YT, Tsai WC, Lin WZ, Wu CC, Yu JC, Tseng VS, Liao GS, Hu JM, Hsu HM, Chang YJ, Lin MC, Chu CM, Yang CY. A Novel IGLC2 Gene Linked With Prognosis of Triple-Negative Breast Cancer. Front Oncol 2022; 11:759952. [PMID: 35155184 PMCID: PMC8829566 DOI: 10.3389/fonc.2021.759952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Immunoglobulin-related genes are associated with the favorable prognosis of triple-negative breast cancer (TNBC) patients. We aimed to analyze the function and prognostic value of immunoglobulin lambda constant 2 (IGLC2) in TNBC patients. METHODS We knocked down the gene expression of IGLC2 (IGLC2-KD) in MDA-MB-231 cells to evaluate the proliferation, migration, and invasion of tumors via 3-(4,5-Dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, wound healing, and transwell cell migration assay respectively. Relapse-free survival (RFS) and distant metastasis-free survival (DMFS) analyses were conducted using the KM plotter online tool. The GSE76275 data set was used to analyze the association of IGLC2 and clinical characteristics. A pathway enrichment analysis was conducted using the next-generation sequencing data of wild-type and IGLC2-KD MDA-MB-231 cells. RESULTS The low gene expression of IGLC2 was related to unfavorable RFS, DMFS. The high expression of IGLC2 was exhibited in the basal-like immune-activated (BLIA) TNBC molecular subtype, which was immune-activated and showed excellent response to immune therapy. IGLC2 was positively correlated with programmed death-ligand 1 (PD-L1) as shown by Spearman correlation (r = 0.25, p < 0.0001). IGLC2 had a strong prognostic effect on lymph node-negative TNBC (RFS range: 0.31, q value= 8.2e-05; DMFS = 0.16, q value = 8.2e-05) but had no significance on lymph node-positive ones. The shRNA-mediated silencing of IGLC2 increased the proliferation, migration, and invasion of MDA-MB-231 cells. The results of pathway enrichment analysis showed that IGLC2 is related to the PI3K-Akt signaling pathway, MAPK signaling pathway, and extracellular matrix-receptor interaction. We confirmed that MDA-MB-231 tumor cells expressed IGLC2, subverting the traditional finding of generation by immune cells. CONCLUSIONS IGLC2 linked with the proliferation, migration, and invasion of MDA-MB-231 cells. A high expression of IGLC2 was related to favorable prognosis for TNBC patients. IGLC2 may serve as a biomarker for the identification of TNBC patients who can benefit the most from immune checkpoint blockade treatment.
Collapse
Affiliation(s)
- Yu-Tien Chang
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Zhi Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Chao Wu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jyh-Cherng Yu
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Vincent S. Tseng
- Department of Computer Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Guo-Shiou Liao
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Je-Ming Hu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Huan-Ming Hsu
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Surgery, Songshan Branch of Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Jia Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Cancer Research Center and Translational Laboratory, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Meng-Chiung Lin
- Division of Gastroenterology, Department of Medicine, Taichung Armed Forces General Hospital, Taichung, Taiwan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Chi-Ming Chu
- Division of Biostatistics and Informatics, Department of Epidemiology, School of Public Health, National Defense Medical Center, Taipei, Taiwan
- Big Data Research Center, Fu-Jen Catholic University, New Taipei City, Taiwan
- Department of Public Health, China Medical University, Taichung, Taiwan
- Department of Healthcare Administration and Medical Informatics College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Yi Yang
- Department of Surgery, Songshan Branch of Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
12
|
Huang S, Huang P, Wu H, Wang S, Liu G. LINC02381 aggravates breast cancer through the miR-1271-5p/FN1 axis to activate PI3K/AKT pathway. Mol Carcinog 2021; 61:346-358. [PMID: 34882856 DOI: 10.1002/mc.23375] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/27/2021] [Accepted: 11/12/2021] [Indexed: 01/23/2023]
Abstract
Emerging investigations have demonstrated that lncRNAs are key crucial modulators in cancer. In this study, we investigated the role of LINC02381 in breast cancer (BC). Reverse transcriptase quantitative polymerase chain reaction measured the LINC02381 level in BC tissues and cells. Colony formation, EdU staining, wound healing and Transwell experiments examined the impact of LINC02381 depletion on BC cell phenotypes. Relationship among miR-1271-5p, LINC02381, and FN1 was tested through applying RIP, luciferase reporter, and RNA pull-down assays. We found that LINC02381 expression was elevated in BC. Functionally, LINC02381 knockdown hampered BC cell proliferation, migration, and invasion. LINC02381 overexpression accelerated tumor formation in vivo. Mechanistically, LINC02381 acted as a ceRNA to increase FN1 via decoying miR-1271-5p. Additionally, LINC02381 activated PI3K/AKT pathway by upregulating FN1. Rescue assays indicated that FN1 upregulation or PI3K/AKT activation rescued the LINC02381 knockdown-mediated inhibition on malignant phenotypes of BC cells. Overall, LINC02381 exerts carcinogenic effects in BC by the miR-1271-5p/FN1 axis to activate PI3K/AKT pathway.
Collapse
Affiliation(s)
- Shoucheng Huang
- College of Life and Health Science, Anhui Science and Technology University, Fengyang, Anhui, China
| | - Ping Huang
- College of Chemical and Materials Engineering, Anhui Science and Technology University, Fengyang, Anhui, China
| | - Huazhang Wu
- School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui, China
| | - Song Wang
- College of Life and Health Science, Anhui Science and Technology University, Fengyang, Anhui, China
| | - Guodong Liu
- College of Life and Health Science, Anhui Science and Technology University, Fengyang, Anhui, China
| |
Collapse
|
13
|
Anuraga G, Wang WJ, Phan NN, An Ton NT, Ta HDK, Berenice Prayugo F, Minh Xuan DT, Ku SC, Wu YF, Andriani V, Athoillah M, Lee KH, Wang CY. Potential Prognostic Biomarkers of NIMA (Never in Mitosis, Gene A)-Related Kinase (NEK) Family Members in Breast Cancer. J Pers Med 2021; 11:1089. [PMID: 34834441 PMCID: PMC8625415 DOI: 10.3390/jpm11111089] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
Breast cancer remains the most common malignant cancer in women, with a staggering incidence of two million cases annually worldwide; therefore, it is crucial to explore novel biomarkers to assess the diagnosis and prognosis of breast cancer patients. NIMA-related kinase (NEK) protein kinase contains 11 family members named NEK1-NEK11, which were discovered from Aspergillus Nidulans; however, the role of NEK family genes for tumor development remains unclear and requires additional study. In the present study, we investigate the prognosis relationships of NEK family genes for breast cancer development, as well as the gene expression signature via the bioinformatics approach. The results of several integrative analyses revealed that most of the NEK family genes are overexpressed in breast cancer. Among these family genes, NEK2/6/8 overexpression had poor prognostic significance in distant metastasis-free survival (DMFS) in breast cancer patients. Meanwhile, NEK2/6 had the highest level of DNA methylation, and the functional enrichment analysis from MetaCore and Gene Set Enrichment Analysis (GSEA) suggested that NEK2 was associated with the cell cycle, G2M checkpoint, DNA repair, E2F, MYC, MTORC1, and interferon-related signaling. Moreover, Tumor Immune Estimation Resource (TIMER) results showed that the transcriptional levels of NEK2 were positively correlated with immune infiltration of B cells and CD4+ T Cell. Collectively, the current study indicated that NEK family genes, especially NEK2 which is involved in immune infiltration, and may serve as prognosis biomarkers for breast cancer progression.
Collapse
Affiliation(s)
- Gangga Anuraga
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (G.A.); (H.D.K.T.); (K.-H.L.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (F.B.P.); (D.T.M.X.); (S.-C.K.)
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia;
| | - Wei-Jan Wang
- Research Center for Cancer Biology, Department of Biological Science and Technology, China Medical University, Taichung 40604, Taiwan;
| | - Nam Nhut Phan
- Institute for Environmental Science, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Vietnam; (N.N.P.); (N.T.A.T.)
| | - Nu Thuy An Ton
- Institute for Environmental Science, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Vietnam; (N.N.P.); (N.T.A.T.)
| | - Hoang Dang Khoa Ta
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (G.A.); (H.D.K.T.); (K.-H.L.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (F.B.P.); (D.T.M.X.); (S.-C.K.)
| | - Fidelia Berenice Prayugo
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (F.B.P.); (D.T.M.X.); (S.-C.K.)
| | - Do Thi Minh Xuan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (F.B.P.); (D.T.M.X.); (S.-C.K.)
| | - Su-Chi Ku
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (F.B.P.); (D.T.M.X.); (S.-C.K.)
| | - Yung-Fu Wu
- Department of Medical Research, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Vivin Andriani
- Department of Biological Science, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia;
| | - Muhammad Athoillah
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia;
| | - Kuen-Haur Lee
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (G.A.); (H.D.K.T.); (K.-H.L.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (F.B.P.); (D.T.M.X.); (S.-C.K.)
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Yang Wang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (G.A.); (H.D.K.T.); (K.-H.L.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (F.B.P.); (D.T.M.X.); (S.-C.K.)
| |
Collapse
|
14
|
Ho PJ, Dorajoo R, Ivanković I, Ong SS, Khng AJ, Tan BKT, Tan VKM, Lim SH, Tan EY, Tan SM, Tan QT, Yan Z, Ngeow J, Sim Y, Chan P, Chuan JCJ, Chan CW, Tang SW, Hartman M, Li J. DNA methylation and breast cancer-associated variants. Breast Cancer Res Treat 2021; 188:713-727. [PMID: 33768416 DOI: 10.1007/s10549-021-06185-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND A breast cancer polygenic risk score (PRS) comprising 313 common variants reliably predicts disease risk. We examined possible relationships between genetic variation, regulation, and expression to clarify the molecular alterations associated with these variants. METHODS Genome-wide methylomic variation was quantified (MethylationEPIC) in Asian breast cancer patients (1152 buffy coats from peripheral whole blood). DNA methylation (DNAm) quantitative trait loci (mQTL) mapping was performed for 235 of the 313 variants with minor allele frequencies > 5%. Stability of identified mQTLs (p < 5e-8) across lifetime was examined using a public mQTL database. Identified mQTLs were also mapped to expression quantitative trait loci (eQTLs) in the Genotype-Tissue Expression Project and the eQTLGen Consortium. RESULTS Breast cancer PRS was not associated with DNAm. A higher proportion of significant cis-mQTLs were observed. Of 822 significant cis-mQTLs (179 unique variants) identified in our dataset, 141 (59 unique variants) were significant (p < 5e-8) in a public mQTL database. Eighty-six percent (121/141) of the matched mQTLs were consistent at multiple time points (birth, childhood, adolescence, pregnancy, middle age, post-diagnosis, or treatment). Ninety-three variants associated with DNAm were also cis-eQTLs (35 variants not genome-wide significant). Multiple loci in the breast cancer PRS are associated with DNAm, contributing to the polygenic nature of the disease. These mQTLs are mostly stable over time. CONCLUSIONS Consistent results from DNAm and expression data may reveal new candidate genes not previously associated with breast cancer.
Collapse
Affiliation(s)
- Peh Joo Ho
- Genome Institute of Singapore, Human Genetics, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Human Genetics, Singapore, Singapore
- Health Systems and Services Research, Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Ivna Ivanković
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Biomedical Informatics, University Hospital of Zurich, Zurich, Switzerland
| | - Seeu Si Ong
- Genome Institute of Singapore, Human Genetics, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | | | - Benita Kiat-Tee Tan
- Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Veronique Kiak Mien Tan
- Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Swee Ho Lim
- KK Breast Department, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Ern Yu Tan
- Department of General Surgery, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | - Su-Ming Tan
- Division of Breast Surgery, Changi General Hospital, Singapore, Singapore
| | - Qing Ting Tan
- KK Breast Department, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Zhiyan Yan
- KK Breast Department, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Joanne Ngeow
- Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, Singapore
- Cancer Genetics Service, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Clinical Program, Duke NUS, Singapore, Singapore
| | - Yirong Sim
- Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Patrick Chan
- Department of General Surgery, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | | | - Ching Wan Chan
- Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
| | - Siau Wei Tang
- Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
| | - Mikael Hartman
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
| | - Jingmei Li
- Genome Institute of Singapore, Human Genetics, Singapore, Singapore.
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore.
| |
Collapse
|
15
|
Chen S, Chen Y, Wen Y, Cai W, Zhu P, Yuan W, Li Y, Fan X, Wan Y, Li F, Zhuang J, Jiang Z, Wu X, Wang Y. miR-590-5p targets RMND5A and promotes migration in pancreatic adenocarcinoma cell lines. Oncol Lett 2021; 22:532. [PMID: 34079591 PMCID: PMC8156640 DOI: 10.3892/ol.2021.12793] [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: 09/29/2020] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Required for meiotic nuclear division 5 homolog A (RMND5A) functions as an E3 ubiquitin ligase. To date, few studies have investigated the role of RMND5A in cancer. In the present study, the expression levels of RMND5A in multiple types of cancer were analyzed using the Gene Expression Profiling Interactive Analysis platform. The results revealed that RMND5A was highly expressed and associated with overall survival in patients with pancreatic adenocarcinoma (PAAD). A wound-healing assay revealed that RMND5A overexpression significantly increased cell migration in the PAAD cell lines AsPC-1 and PANC-1. In silico analysis predicted that RMND5A was a potential target of microRNA(miR)-590-5p. Further in vitro experiments demonstrated that overexpression of miR-590-5p downregulated the expression levels of RMND5A and decreased the migratory ability of the AsPC-1 and PANC-1 cell lines. In addition, overexpression of miR-590-5p attenuated the promoting effects of RMND5A on the migration of AsPC-1 and PANC-1 cells. The results of the present study may further elucidate the mechanisms underlying PAAD progression and provide novel targets for the treatment of PAAD.
Collapse
Affiliation(s)
- Sixing Chen
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yu Chen
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yao Wen
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Wanwan Cai
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, P.R. China
| | - Wuzhou Yuan
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yongqing Li
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiongwei Fan
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yongqi Wan
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Fang Li
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Jian Zhuang
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, P.R. China
| | - Zhigang Jiang
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiushan Wu
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yuequn Wang
- The Center for Heart Development, State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| |
Collapse
|
16
|
Li KZ, Yin YX, Tang YP, Long L, Xie MZ, Li JL, Ding K, Hu BL. Construction of a long noncoding RNA-based competing endogenous RNA network and prognostic signatures of left- and right-side colon cancer. Cancer Cell Int 2021; 21:211. [PMID: 33858429 PMCID: PMC8048080 DOI: 10.1186/s12935-021-01901-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 03/30/2021] [Indexed: 01/26/2023] Open
Abstract
Background Cancers located on the right and left sides of the colon have distinct clinical and molecular characteristics. This study aimed to explore the regulatory mechanisms of location-specific long noncoding RNAs (lncRNAs) as competing endogenous RNAs (ceRNAs) in colon cancer and identify potential prognostic biomarkers. Method Differentially expressed lncRNAs (DELs), miRNAs (DEMs), and genes (DEGs) between right- and left-side colon cancers were identified by comparing RNA sequencing profiles. Functional enrichment analysis was performed for the DEGs, and a ceRNA network was constructed. Associations between DELs and patient survival were examined, and a DEL-based signature was constructed to examine the prognostic value of these differences. Clinical colon cancer tissues and Gene Expression Omnibus (GEO) datasets were used to validate the results. Results We identified 376 DELs, 35 DEMs, and 805 DEGs between right- and left-side colon cancers. The functional enrichment analysis revealed the functions and pathway involvement of DEGs. A ceRNA network was constructed based on 95 DEL–DEM–DEG interactions. Three DELs (LINC01555, AC015712, and FZD10-AS1) were associated with the overall survival of patients with colon cancer, and a prognostic signature was established based on these three DELs. High risk scores for this signature indicated poor survival, suggesting that the signature has prognostic value for colon cancer. Examination of clinical colon cancer tissues and GEO dataset analysis confirmed the results. Conclusion The ceRNA regulatory network suggests roles for location-specific lncRNAs in colon cancer and allowed the development of an lncRNA-based prognostic signature, which could be used to assess prognosis and determine treatment strategies in patients with colon cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-01901-3.
Collapse
Affiliation(s)
- Ke-Zhi Li
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Yi-Xin Yin
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Yan-Ping Tang
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Long Long
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Ming-Zhi Xie
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Ji-Lin Li
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Ke Ding
- Department of Radiology, Third Affiliated Hospital of Guangxi Medical University, 13 Dancun Road, Nanning, 530031, Guangxi, China.
| | - Bang-Li Hu
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
17
|
Hao M, Liu W, Ding C, Peng X, Zhang Y, Chen H, Dong L, Liu X, Zhao Y, Chen X, Khatoon S, Zheng Y. Identification of hub genes and small molecule therapeutic drugs related to breast cancer with comprehensive bioinformatics analysis. PeerJ 2020; 8:e9946. [PMID: 33083112 PMCID: PMC7556247 DOI: 10.7717/peerj.9946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is one of the most common malignant tumors among women worldwide and has a high morbidity and mortality. This research aimed to identify hub genes and small molecule drugs for breast cancer by integrated bioinformatics analysis. After downloading multiple gene expression datasets from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, 283 overlapping differentially expressed genes (DEGs) significantly enriched in different cancer-related functions and pathways were obtained using LIMMA, VennDiagram and ClusterProfiler packages of R. We then analyzed the topology of protein–protein interaction (PPI) network with overlapping DEGs and further obtained six hub genes (RRM2, CDC20, CCNB2, BUB1B, CDK1, and CCNA2) from the network via STRING and Cytoscape. Subsequently, we conducted genes expression verification, genetic alterations evaluation, immune infiltration prediction, clinicopathological parameters analysis, identification of transcriptional and post-transcriptional regulatory molecules, and survival analysis for these hub genes. Meanwhile, 29 possible drug candidates (e.g., Cladribine, Gallium nitrate, Alvocidib, 1β-hydroxyalantolactone, Berberine hydrochloride, Nitidine chloride) were identified from the DGIdb database and the GSE85871 dataset. In addition, some transcription factors and miRNAs (e.g., E2F1, PTTG1, TP53, ZBTB16, hsa-miR-130a-3p, hsa-miR-204-5p) targeting hub genes were identified as key regulators in the progression of breast cancer. In conclusion, our study identified six hub genes and 29 potential drug candidates for breast cancer. These findings may advance understanding regarding the diagnosis, prognosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Mingqian Hao
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Wencong Liu
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Chuanbo Ding
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Xiaojuan Peng
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Yue Zhang
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Huiying Chen
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Ling Dong
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Xinglong Liu
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Yingchun Zhao
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Xueyan Chen
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Sadia Khatoon
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Yinan Zheng
- School of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| |
Collapse
|
18
|
Wu Z, Wang W, Wang Y, Wang X, Sun S, Yao Y, Zhang Y, Ren Z. Long noncoding RNA LINC00963 promotes breast cancer progression by functioning as a molecular sponge for microRNA-625 and thereby upregulating HMGA1. Cell Cycle 2020; 19:610-624. [PMID: 32052688 PMCID: PMC7100992 DOI: 10.1080/15384101.2020.1728024] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/28/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Extensive research has shown that LINC00963 is aberrantly expressed in human cancers, and that dysregulation of LINC00963 is implicated in the initiation and progression of human cancers. The expression and functions of LINC00963 in breast cancer are still unclear. Our aims were to measure the expression of LINC00963 in breast cancer, determine its effects on malignant behaviors of tumor cells, and uncover the molecular events underlying the actions of LINC00963 in breast cancer. Herein, LINC00963 was found to be overexpressed in breast cancer samples, and its overexpression was correlated with lymph node metastasis, TNM stage and differentiation grade. Patients with breast cancer harboring higher LINC00963 expression showed shorter overall survival than did the patients with lower LINC00963 expression. Functional experiments revealed that depletion of LINC00963 inhibited breast cancer cell proliferation, migration, and invasion and facilitated apoptosis in vitro and impaired tumor growth in vivo. Mechanism investigation revealed that LINC00963 can interact with microRNA-625 (miR-625). LINC00963 worked as a competitive endogenous RNA for miR-625 to weaken the suppressive effect of miR-625 on high mobility group AT-hook 1 (HMGA1) in breast cancer cells. Furthermore, miR-625 inhibition and HMGA1 restoration both abrogated the effects of LINC00963 silencing on breast cancer cells. Our findings indicate that the LINC00963-miR-625-HMGA1 pathway plays an important role in the malignancy of breast cancer in vitro and in vivo. Hence, targeting this pathway may be a novel strategy against breast cancer.
Collapse
Affiliation(s)
- Zhen Wu
- Department of Breast and Thyroid Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, P.R. China
| | - Wei Wang
- Department of Breast and Thyroid Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, P.R. China
| | - Yongkun Wang
- Department of Breast and Thyroid Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, P.R. China
| | - Xin Wang
- Department of Breast and Thyroid Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, P.R. China
| | - Shanping Sun
- Department of Breast and Thyroid Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, P.R. China
| | - Yumin Yao
- Department of Breast and Thyroid Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, P.R. China
| | - Yang Zhang
- Department of Breast and Thyroid Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, P.R. China
| | - Zhongxi Ren
- Department of Breast and Thyroid Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, P.R. China
| |
Collapse
|
19
|
The CTLH Complex in Cancer Cell Plasticity. JOURNAL OF ONCOLOGY 2019; 2019:4216750. [PMID: 31885576 PMCID: PMC6907057 DOI: 10.1155/2019/4216750] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/24/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022]
Abstract
Cancer cell plasticity is the ability of cancer cells to intermittently morph into different fittest phenotypic states. Due to the intrinsic capacity to change their composition and interactions, protein macromolecular complexes are the ideal instruments for transient transformation. This review focuses on a poorly studied mammalian macromolecular complex called the CTLH (carboxy-terminal to LisH) complex. Currently, this macrostructure includes 11 known members (ARMC8, GID4, GID8, MAEA, MKLN1, RMND5A, RMND5B, RANBP9, RANBP10, WDR26, and YPEL5) and it has been shown to have E3-ligase enzymatic activity. CTLH proteins have been linked to all fundamental biological processes including proliferation, survival, programmed cell death, cell adhesion, and migration. At molecular level, the complex seems to interact and intertwine with key signaling pathways such as the PI3-kinase, WNT, TGFβ, and NFκB, which are key to cancer cell plasticity. As a whole, the CTLH complex is overexpressed in the most prevalent types of cancer and may hold the key to unlock many of the biological secrets that allow cancer cells to thrive in harsh conditions and resist antineoplastic therapy.
Collapse
|
20
|
Li W, Jin X, Zhao Y, Dai J, Cai Y. Long noncoding RNA GAS6-AS2 sponges microRNA-493, thereby enhancing the malignant characteristics of breast cancer cells via upregulation of FUT4. Pathol Res Pract 2019; 216:152772. [PMID: 31839366 DOI: 10.1016/j.prp.2019.152772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/16/2019] [Accepted: 11/27/2019] [Indexed: 10/25/2022]
Abstract
Long noncoding RNA (lncRNA) GAS6-AS2 serves as an oncogenic lncRNA in various types of human cancer. In this study, we attempted to examine the functions of GAS6-AS2 in breast cancer (BC) and explore the potential mechanisms involved. Reverse-transcription quantitative PCR was carried out to determine GAS6-AS2 expression in BC tissues and cell lines. Multiple functional experiments, including a Cell Counting Kit-8 assay, Transwell migration and invasion assays, and an in vivo nude-mouse xenograft experiment, were conducted to evaluate the effects of GAS6-AS2 in BC cells. GAS6-AS2 expression was high in BC tumors, manifesting a strong correlation with tumor size, lymph node metastasis, TNM stage, and shorter overall survival in patients with BC. A knockdown of GAS6-AS2 restricted BC cell proliferation, migration, and invasion in vitro and retarded tumor growth in vivo. With regard to its mechanism, GAS6-AS2 acted as a competing endogenous RNA that sponged microRNA-493 (miR-493), thereby increasing the expression of fucosyltransferase IV (FUT4). Either miR-493 inhibition or FUT4 upregulation abrogated the consequences of GAS6-AS2 knockdown in BC cells. These results revealed that GAS6-AS2 sponges miR-493 to enhance the malignant characteristics of BC in vitro and in vivo by increasing FUT4 expression. Thus, this lncRNA is an effective therapeutic target in BC and a promising diagnostic biomarker of this cancer.
Collapse
Affiliation(s)
- Wanfeng Li
- Department of Breast Surgery, Jilin Tumor Hospital, Jilin, 130012, PR China
| | - Xintian Jin
- Department of Thoracic Neoplasms, Jilin Tumor Hospital, Jilin, 130012, PR China
| | - Yueming Zhao
- Department of Oncology, Jilin Tumor Hospital, Jilin, 130012, PR China
| | - Jixin Dai
- Department of Oncology, Jilin Tumor Hospital, Jilin, 130012, PR China
| | - Yong Cai
- Department of Breast Surgery, Jilin Tumor Hospital, Jilin, 130012, PR China.
| |
Collapse
|
21
|
Si H, Chen P, Li H, Wang X. Long non-coding RNA H19 regulates cell growth and metastasis via miR-138 in breast cancer. Am J Transl Res 2019; 11:3213-3225. [PMID: 31217890 PMCID: PMC6556666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/13/2019] [Indexed: 06/09/2023]
Abstract
Breast cancer is one of the most common cancers among women. Long non-coding RNAs (lncRNAs) are involved in the initiation and development of breast cancer and lncRNA H19 is a potential oncogenic factor; however, the underlying mechanisms remain unknown. In the present study, the regulatory functions of H19 in breast cancer were investigated. We found that H19 was upregulated in breast cancer tissues and cells and associated with poor prognosis. MiR-138 was downregulated in breast cancer tissues and negatively correlated with the expression of H19 and SOX4. Furthermore, SOX4 was upregulated in breast cancer tissues and positively correlated with H19. Downregulated H19 suppressed the proliferation, invasion and migration of breast cancer cells, but promoted cell cycle arrest and apoptosis. Additionally, miR-138 was identified as a direct target of H19 and SOX4; overexpression of miR-138 inhibited the proliferation, invasion and migration of MDA-MB-231 and MCF-7 cells, but promoted apoptosis, which were abrogated by SOX4 overexpression. Downregulated miR-138 induced cell proliferation, invasion and migration, but inhibited apoptosis of MDA-MB-231 and MCF-7 cells, which were promoted by SOX4 overexpression. In addition, miR-138 overexpression reversed the effects of H19 in breast cancer cells; silencing of H19 inhibited tumor growth and downregulate EMT markers in vivo. In summary, H19 was upregulated in breast cancer and associated with poor prognosis. Silencing of H19 inhibited cell proliferation, invasion and migration, but induced cell cycle arrest and apoptosis by regulating miR-138 and SOX4 in breast cancer.
Collapse
Affiliation(s)
- Haiyan Si
- Department of Breast and Thyroid Surgery, First People’s Hospital of Jiaozuo CityJiaozuo 454000, China
| | - Ping Chen
- Department of Pharmacy, Affiliated Hospital of Shandong Medical CollegeJinan 276000, Shandong, China
| | - Hongtao Li
- Department of Breast and Thyroid Surgery, First People’s Hospital of Jiaozuo CityJiaozuo 454000, China
| | - Xiang Wang
- Physical Examination Centre, The Third Affiliated Hospital of Chongqing Medical UniversityChongqing 401120, China
| |
Collapse
|
22
|
Hou S, Tan J, Yang B, He L, Zhu Y. Effect of alkylglycerone phosphate synthase on the expression profile of circRNAs in the human thyroid cancer cell line FRO. Oncol Lett 2018; 15:7889-7899. [PMID: 29731907 PMCID: PMC5920571 DOI: 10.3892/ol.2018.8356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 02/27/2018] [Indexed: 12/17/2022] Open
Abstract
Thyroid cancer is a common primary tumor in China. Therefore, it is important to investigate the underlying molecular mechanism of thyroid cancer in order to achieve effective individualized treatments. In our previous study, a positive correlation between the expression of alkylglycerone phosphate synthase (AGPS) and the malignant phenotype of thyroid cancer cell lines was identified. The inactivation of AGPS was able to decrease the malignancy of cancer, and inhibit tumor growth and invasion. However, the function of AGPS on thyroid cancer was unclear. In the present study, it was revealed that AGPS was able to regulate the expression of circular RNAs (circRNAs), which may be the mechanism of its anticancer activity. Therefore, the effects of AGPS silencing and knockout on circRNA expression in the thyroid cancer cell line FRO were investigated using circRNAs microarray, and Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed in order to investigate the underlying molecular mechanism of AGPS for the regulation of thyroid cancer through circRNAs.
Collapse
Affiliation(s)
- Shasha Hou
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Graduate School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jian Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Bing Yang
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Lu He
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yu Zhu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vessels and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| |
Collapse
|