1
|
Pan X, Liu J, Zhang Y, Sun C, Li Y, Guo H. Characterization of microRNA-223-3p as a novel promoter of cell proliferation and invasion in papillary thyroid carcinoma. J Cell Commun Signal 2025; 19:e12057. [PMID: 39712860 PMCID: PMC11658962 DOI: 10.1002/ccs3.12057] [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: 05/09/2024] [Revised: 09/23/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
Papillary thyroid carcinoma (PTC), the most common thyroid cancer, has been linked to various molecular alterations. This study focuses on microRNA-223-3p, whose upregulated expression in PTC tissues appears to enhance tumor growth and cellular dysfunctions. Our findings demonstrate that microRNA-223-3p significantly promotes cell proliferation, invasion, and migration and induces epithelial-mesenchymal transition (EMT). Additionally, neurofibromatosis type 2 (NF2) is identified as a direct target, suggesting that microRNA-223-3p could be crucial in PTC pathogenesis and may offer a target for therapeutic intervention.
Collapse
Affiliation(s)
- Xinghe Pan
- Department of General SurgeryCentral Hospital Affiliated to Shenyang Medical CollegeShenyangLiaoningChina
| | - Junliang Liu
- Department of General SurgeryCentral Hospital Affiliated to Shenyang Medical CollegeShenyangLiaoningChina
| | - Yitong Zhang
- Department of General SurgeryCentral Hospital Affiliated to Shenyang Medical CollegeShenyangLiaoningChina
| | - Chenglin Sun
- Department of General SurgeryCentral Hospital Affiliated to Shenyang Medical CollegeShenyangLiaoningChina
| | - You Li
- Department of General SurgeryCentral Hospital Affiliated to Shenyang Medical CollegeShenyangLiaoningChina
| | - Hongpeng Guo
- Department of General SurgeryCentral Hospital Affiliated to Shenyang Medical CollegeShenyangLiaoningChina
| |
Collapse
|
2
|
Peng H, Zhu Z, Xing J, Xu Q, Man C, Wang S, Liu Y, Zhang Z. Expression profiling and bioinformatics analysis of serum exosomal circular RNAs in lymph node metastasis of papillary thyroid carcinoma. J Biomed Res 2024; 39:1-15. [PMID: 38812291 PMCID: PMC11982685 DOI: 10.7555/jbr.37.20230304] [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: 12/21/2023] [Revised: 04/28/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024] Open
Abstract
Most papillary thyroid carcinoma (PTC) patients have a good prognosis, but lymph node metastasis (LNM) is the most common progressive manifestation and often leads to a poor-prognosis. However, few studies focused on the underlying mechanisms of LNM. This study aimed to identity the potential role of exosomal circRNAs that contribute to LNM in PTC. We found that 9000 aberrantly expressed exosomal circRNAs in PTC patients with LNM, including 684 observably upregulation and 2193 notably downregulation. Functional enrichment analyses indicated that these aberrantly expressed circRNAs were mainly enriched in a variety of molecules and signaling pathways related to the progression and LNM of PTC. Bioinformatics analysis screened 14 circRNA-miRNA-mRNA networks associated with LNM-related signaling pathways in PTC. Moreover, circTACC2-miR-7-EGFR and circBIRC6-miR-24-3p-BCL2L11 axes were verified for potential involvement in PTC with LNM. Additionally, 4 upregulated circRNAs-related hub genes and 8 hub genes associated with downregulated circRNAs were screened, some of which were involved in LNM of PTC through verification. Collectively, our data provided a novel framework for in-depth investigation of the function of dysregulated exosomal circRNAs and their potential biomarkers in PTC patients with LNM.
Collapse
Affiliation(s)
- Huiyong Peng
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Laboratory Medicine, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, Jiangsu 212002, China
| | - Zhangwei Zhu
- Department of Endocrinology, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, Jiangsu 212002, China
| | - Jie Xing
- Department of Laboratory Medicine, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, Jiangsu 212002, China
| | - Qian Xu
- Department of Endocrinology, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, Jiangsu 212002, China
| | - Changfeng Man
- Department of Oncology, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, Jiangsu 212002, China
| | - Shengjun Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212008, China
| | - Yingzhao Liu
- Department of Endocrinology, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, Jiangsu 212002, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
3
|
Chen X, Zhang T, He YQ, Miao TW, Yin J, Ding Q, Yang M, Chen FY, Zeng HP, Liu J, Zhu Q. NGEF is a potential prognostic biomarker and could serve as an indicator for immunotherapy and chemotherapy in lung adenocarcinoma. BMC Pulm Med 2024; 24:248. [PMID: 38764064 PMCID: PMC11102621 DOI: 10.1186/s12890-024-03046-1] [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: 10/16/2023] [Accepted: 05/06/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Neuronal guanine nucleotide exchange factor (NGEF) plays a key role in several cancers; however, its role in lung adenocarcinoma (LUAD) remains unclear. The aim of this study was to evaluate the efficacy of NGEF as a prognostic biomarker and potential therapeutic target for LUAD. METHODS NGEF expression data for multiple cancers and LUAD were downloaded from multiple databases. The high- and low-NGEF expression groups were constructed based on median NGEF expression in LUAD samples, and then performed Kaplan-Meier survival analysis. Differentially expressed genes (DEGs) from the two NGEF expression groups were screened and applied to construct a protein-protein interaction network. The primary pathways were obtained using gene set enrichment analysis. The associations between NGEF expression and clinical characteristics, immune infiltration, immune checkpoint inhibitors (ICIs), sensitivity to chemotherapy, and tumor mutation burden (TMB) were investigated using R. Levels of NGEF expression in the lung tissue was validated using single-cell RNA sequencing, quantitative polymerase chain reaction (qPCR), immunohistochemical staining, and western blot analysis. RESULTS The expression of NGEF mRNA was upregulated in multiple cancers. mRNA and protein expression levels of NGEF were higher in patients with LUAD than in controls, as validated using qPCR and western blot. High NGEF expression was an independent prognostic factor for LUAD and was associated with advanced tumor stage, large tumor size, more lymph node metastasis, and worse overall survival (OS). A total of 182 overlapping DEGs were screened between The Cancer Genome Atlas and GSE31210, among which the top 20 hub genes were identified. NGEF expression was mainly enriched in the pathways of apoptosis, cell cycle, and DNA replication. Moreover, elevated NGEF expression were associated with a high fraction of activated memory CD4+ T cells and M0 macrophages; elevated expression levels of the ICIs: programmed cell death 1 and programmed cell death 1 ligand 1 expression; higher TMB; and better sensitivity to bortezomib, docetaxel, paclitaxel, and parthenolide, but less sensitivity to axitinib and metformin. CONCLUSION NGEF expression is upregulated in LUAD and is significantly associated with tumor stages, OS probability, immune infiltration, immunotherapy response, and chemotherapy response. NGEF may be a potential diagnostic and prognostic biomarker and therapeutic target in LUAD.
Collapse
Affiliation(s)
- Xin Chen
- Department of Integrated Traditional Chinese and Western Medicine, Zigong First People's Hospital, Zigong, China.
| | - Tao Zhang
- Department of Intensive Care Unit, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Yan-Qiu He
- Department of Integrated Traditional Chinese and Western Medicine, Zigong First People's Hospital, Zigong, China
| | - Ti-Wei Miao
- Department of Integrated Traditional Chinese and Western Medicine, Zigong First People's Hospital, Zigong, China
| | - Jie Yin
- School of Automation & Information Engineering, Sichuan university of Science & Engineering, Zigong, China
| | - Qian Ding
- Department of Integrated Traditional Chinese and Western Medicine, Zigong First People's Hospital, Zigong, China
| | - Mei Yang
- Department of Integrated Traditional Chinese and Western Medicine, Zigong First People's Hospital, Zigong, China
| | - Fang-Ying Chen
- Department of Tuberculosis, The Third People's Hospital of Tibet Autonomous Region, Lhasa, China
| | - Hong-Ping Zeng
- Department of Integrated Traditional Chinese and Western Medicine, Zigong First People's Hospital, Zigong, China
| | - Jie Liu
- Department of Integrated Traditional Chinese and Western Medicine, Zigong First People's Hospital, Zigong, China
| | - Qi Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Zigong First People's Hospital, Zigong, China
| |
Collapse
|
4
|
He T, Sun X, Wu C, Yao L, Zhang Y, Liu S, Jiang Y, Li Y, Wang M, Xu Y. PROS1, a clinical prognostic biomarker and tumor suppressor, is associated with immune cell infiltration in breast cancer: A bioinformatics analysis combined with experimental verification. Cell Signal 2023; 112:110918. [PMID: 37827342 DOI: 10.1016/j.cellsig.2023.110918] [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: 06/09/2023] [Revised: 09/12/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND PROS1 is an encoding gene that can generate protein S. This protein is a glycoprotein found in plasma that conducts physiological functions with vitamin K. However, the impact of its expression remains absent in the progression and prognosis of breast cancer (BC). METHODS In this study, we comprehensively explored the expression of PROS1 in BC and its relationship with BC patient survival, prognosis, and other clinicopathological features. We investigated how PROS1 influenced the malignant biological behavior of BC cells. A series of enrichment analyses were conducted, and the immune landscape was explored in BC affected by PROS1. We also determined correlations between PROS1 and common drug sensitivities used for BC treatments. RESULTS PROS1 had low expression in BC, which tended to result in poor survival of BC patients. Overexpressed PROS1 inhibited the migration and invasion of BC cells as well as the epithelial-mesenchymal transition process by downregulating SNAIL. Functional enrichment analyses revealed that PROS1 was more active in extracellular matrix (ECM) organization and structural constituent, ECM-receptor interaction, and other pathways with its related genes. PROS1 was also found to affect immune activity, including various immune cells infiltrating BC. BC patients with high PROS1 expression tended to have lower IC50 values of three common medications and obtained better efficacy. CONCLUSIONS PROS1 can become a promising prognostic factor and a possible therapeutic target in BC patients and suppress BC cell metastatic potential. In addition, PROS1 is a crucial factor in immune infiltration in BC.
Collapse
Affiliation(s)
- Tianyi He
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xiangyu Sun
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang 110001, China
| | - Chen Wu
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang 110001, China
| | - Litong Yao
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yingfan Zhang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang 110001, China
| | - Shiyang Liu
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yuhan Jiang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, PKU International Cancer Institute, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Yixiao Li
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, PKU International Cancer Institute, MOE Key Laboratory of Carcinogenesis and Translational Research and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Mozhi Wang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang 110001, China
| | - Yingying Xu
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
5
|
Xiao H, Wang G, Zhao M, Shuai W, Ouyang L, Sun Q. Ras superfamily GTPase activating proteins in cancer: Potential therapeutic targets? Eur J Med Chem 2023; 248:115104. [PMID: 36641861 DOI: 10.1016/j.ejmech.2023.115104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
To search more therapeutic strategies for Ras-mutant tumors, regulators of the Ras superfamily involved in the GTP/GDP (guanosine triphosphate/guanosine diphosphate) cycle have been well concerned for their anti-tumor potentials. GTPase activating proteins (GAPs) provide the catalytic group necessary for the hydrolysis of GTPs, which accelerate the switch by cycling between GTP-bound active and GDP-bound inactive forms. Inactivated GAPs lose their function in activating GTPase, leading to the continuous activation of downstream signaling pathways, uncontrolled cell proliferation, and eventually carcinogenesis. A growing number of evidence has shown the close link between GAPs and human tumors, and as a result, GAPs are believed as potential anti-tumor targets. The present review mainly summarizes the critically important role of GAPs in human tumors by introducing the classification, function and regulatory mechanism. Moreover, we comprehensively describe the relationship between dysregulated GAPs and the certain type of tumor. Finally, the current status, research progress, and clinical value of GAPs as therapeutic targets are also discussed, as well as the challenges and future direction in the cancer therapy.
Collapse
Affiliation(s)
- Huan Xiao
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Min Zhao
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Expression Profile and Diagnostic Significance of MicroRNAs in Papillary Thyroid Cancer. Cancers (Basel) 2022; 14:cancers14112679. [PMID: 35681658 PMCID: PMC9179248 DOI: 10.3390/cancers14112679] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 12/05/2022] Open
Abstract
The incidence of papillary thyroid cancer (PTC) has increased in recent years. To improve the diagnostic management of PTC, we propose the use of microRNAs (miRNAs) as a biomarker. Our aim in this study was to evaluate the miRNA expression pattern in PTC using NanoString technology. We identified ten miRNAs deregulated in PTC compared with reference tissue: miR-146b-5p, miR-221-3p, miR-221-5p, miR-34-5p, miR-551b-3p, miR-152-3p, miR-15a-5p, miR-31-5p, and miR-7-5p (FDR < 0.05; |fold change (FC)| ≥ 1.5). The gene ontology (GO) analysis of differentially expressed miRNA (DEM) target genes identified the predominant involvement of epidermal growth factor receptor (EGFR), tyrosine kinase inhibitor resistance, and pathways in cancer in PTC. The highest area under the receiver operating characteristic (ROC) curve (AUC) for DEMs was found for miR-146-5p (AUC = 0.770) expression, indicating possible clinical applicability in PTC diagnosis. The combination of four miRNAs (miR-152-3p, miR-221-3p, miR-551b-3p, and miR-7-5p) showed an AUC of 0.841. Validation by real-time quantitative polymerase chain reactions (qRT-PCRs) confirmed our findings. The introduction of an miRNA diagnostic panel based on the results of our study may help to improve therapeutic decision making for questionable cases. The use of miRNAs as biomarkers of PTC may become an aspect of personalized medicine.
Collapse
|
7
|
Chen X, Lin S, Lin Y, Wu S, Zhuo M, Zhang A, Zheng J, You Z. BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma. J Transl Med 2022; 20:79. [PMID: 35123502 PMCID: PMC8818187 DOI: 10.1186/s12967-022-03260-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/17/2022] [Indexed: 12/17/2022] Open
Abstract
Abstract
Background
Papillary thyroid carcinoma (PTC) is one of most prevalent malignant endocrine neoplasms, and it is associated with a high frequency of BRAF gene mutations, which lead to lymphatic metastasis and distant metastasis that promote tumor progression. The molecular mechanism of PTC and the role of BRAF mutation in PTC progression and development need to be further elucidated.
Methods
In this study, a comprehensive bioinformatics analysis was performed to identify the differentially expressed genes and signaling pathways in thyroid cancer patients carrying mutant BRAF. Then, we confirmed the prognostic role of WT1 in thyroid cancer patients. Immunohistochemistry was performed to measure the expression profile of WT1 in PTC tissue. Lentivirus shWT1 was transfected into BRAFV600E (mutant) PTC cells to stably inhibit WT1 expression. CCK-8, EdU, immunofluorescence, colony formation, cell migration, cell wound healing, apoptosis and autophagy assays were performed to assess the biological functions of WT1 in BRAFV600E PTC cells. RNA sequencing, immunohistochemistry and immunoblotting were performed to explore the molecular mechanism of WT1 in BRAFV600E PTC cells.
Results
The results confirmed that “epithelial cell proliferation”, “apoptosis” and “selective autophagy” were closely associated with this BRAF mutant in these thyroid cancer patients. Knocking down BRAF-activated WT1 effectively inhibited the proliferation and migration of BRAFV600E PTC cells. Silencing WT1 significantly inhibited autophagy and promoted the apoptosis of BRAFV600E PTC cells. Mechanistic investigations showed that silencing WT1 expression remarkably suppressed the AKT/mTOR and ERK/P65 signaling pathways in BRAFV600E PTC cells.
Conclusion
All these results indicate that WT1 is a promising prognostic biomarker and facilitates PTC progression and development of cells carrying the BRAFV600E mutation.
Collapse
|
8
|
Hong S, Xie Y, Cheng Z, Li J, He W, Guo Z, Zhang Q, Peng S, He M, Yu S, Xu L, Liu R, Xu T, Zhang Y, Li Y, Wang J, Lv W, Yu J, Xiao H. Distinct molecular subtypes of papillary thyroid carcinoma and gene signature with diagnostic capability. Oncogene 2022; 41:5121-5132. [PMID: 36253446 PMCID: PMC9674518 DOI: 10.1038/s41388-022-02499-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 12/24/2022]
Abstract
Papillary thyroid carcinoma (PTC) is heterogeneous and its molecular characteristics remain elusive. We integrated transcriptomic sequencing, genomic analysis and clinicopathologic information from 582 tissue samples of 216 PTC and 75 benign thyroid nodule (BTN) patients. We discovered four subtypes of PTC including Immune-enriched Subtype, BRAF-enriched Subtype, Stromal Subtype and CNV-enriched Subtype. Molecular subtypes were validated in an external cohort of 497 PTC cases from the TCGA. Tumors in the Immune-enriched Subtype showed higher immune infiltration and overexpression of immune checkpoints, whilst BRAF-enriched Subtype showed a higher tendency for extrathyroidal extension and more advanced TNM stage. Key oncogenes including LRRK2, SLC34A2, MUC1, FOXQ1 and KRT19 were overexpressed and enriched in oncogenic MAPK and PI3K/AKT signaling pathways in BRAF-enriched subtype. Further analysis of BRAF-enriched Subtype identified three subclasses with different degrees of malignancies. We also uncovered the molecular link of the initiation and progression from BTN to subtypes of PTC using trajectory analysis. Moreover, a 20-gene expression signature was generated for differential diagnosis of PTC from BTN patients. Together, our work identified previously unreported molecular subtypes of PTC, offering opportunities to stratify patients into optimal treatment plans based on molecular subtyping.
Collapse
Affiliation(s)
- Shubin Hong
- grid.412615.50000 0004 1803 6239Department of endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yubin Xie
- grid.412615.50000 0004 1803 6239Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Cheng
- grid.412615.50000 0004 1803 6239Department of endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jie Li
- grid.412615.50000 0004 1803 6239Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weiman He
- grid.412615.50000 0004 1803 6239Department of endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhuming Guo
- grid.488530.20000 0004 1803 6191Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Quan Zhang
- grid.488530.20000 0004 1803 6191Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sui Peng
- grid.412615.50000 0004 1803 6239Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China ,grid.412615.50000 0004 1803 6239Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Minghui He
- grid.412615.50000 0004 1803 6239Clinical Trials Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuang Yu
- grid.412615.50000 0004 1803 6239Department of endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lixia Xu
- grid.412615.50000 0004 1803 6239Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rengyun Liu
- grid.412615.50000 0004 1803 6239Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tianyi Xu
- grid.412615.50000 0004 1803 6239Department of endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunjian Zhang
- grid.412615.50000 0004 1803 6239Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanbing Li
- grid.412615.50000 0004 1803 6239Department of endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiguang Wang
- grid.24515.370000 0004 1937 1450Division of Life Science, Department of Chemical and Biological Engineering, State Key Laboratory of Molecular Neuroscience and Hong Kong Center for Neurodegenerative Diseases, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Weiming Lv
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Jun Yu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China. .,Institute of Digestive Disease and Department of Medicine & Therapeutics, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Haipeng Xiao
- Department of endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
9
|
Evaluation of the Prognostic Value of Solute Carrier Family 34 Member 2 "SLC34A2" in Papillary Thyroid Carcinoma: An Immunohistochemical Study. ACTA ACUST UNITED AC 2021; 2021:3198555. [PMID: 34336552 PMCID: PMC8298178 DOI: 10.1155/2021/3198555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 12/24/2022]
Abstract
Background Papillary thyroid carcinoma (PTC) usually has an indolent clinical course, yet a subset of patients might show an aggressive course. Thus, better stratification of at-risk patients is mandatory for proper management. Solute carrier family 34 member 2 (SLC34A2) is an independent prognostic indicator in several cancers. However, only a few studies have been conducted to evaluate the prognostic value of SLC34A2 in PTC, with none of them assessing its immunohistochemical (IHC) expression in a large cohort of patients with PTC or exploring its possible relationship with tumor progression. Aim of the Study. We aimed to evaluate the IHC expression of SLC34A2 in a large series of PTC patients, correlate its expression with established clinicopathological factors, and find any possible relationship between this marker and patient prognosis. Material and Methods. A total of 476 samples (including 238 samples of PTC and 238 samples of normal thyroid tissue) collected between 2002 and 2005 were extracted from the archives of the Pathology Lab, Ain Shams University Hospitals. IHC analysis was performed using an anti-SLC34A2 antibody. Follow-up data were obtained. Results High SLC34A2 expression significantly correlated with important adverse clinicopathological parameters of PTC—i.e., late tumor stage, positive extrathyroid extension, tumor size > 4 cm, and age ≥ 55 years (p ≤ 0.001 for each). Kaplan–Meier analysis revealed that high SLC34A2 expression significantly correlated with shorter disease-free survival (DFS; p = 0.005), but not with overall survival (p = 0.111). Multivariate analysis showed SLC34A2 to be an independent prognostic factor affecting DFS. Conclusions High SLC34A2 IHC expression correlated with adverse clinicopathological prognostic parameters. Furthermore, SLC34A2 was identified as an independent factor for DFS that could serve to improve risk stratification of PTC patients for better management.
Collapse
|