1
|
Liu X, Ma Z, Zhang X, Li S, An J, Luo Z. Research Progress of Long Non-coding RNA-ZFAS1 in Malignant Tumors. Cell Biochem Biophys 2024:10.1007/s12013-024-01441-3. [PMID: 39060915 DOI: 10.1007/s12013-024-01441-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Long non-coding RNAs (lncRNAs), although incapable of encoding proteins, play crucial roles in multiple layers of gene expression regulation, epigenetic modifications, and post-transcriptional regulation. Zinc finger antisense 1 (ZFAS1), a lncRNA located in the 20q13 region of the human genome, exhibits dual functions as an oncogene or tumor suppressor in various human malignancies. ZFAS1 plays a crucial role in cancer progression, metastasis, invasion, apoptosis, cell cycle regulation, and drug resistance through complex molecular mechanisms. Additionally, ZFAS1 has a long half-life of over 16 h, demonstrating exceptional stability, and making it a potential biomarker. This review integrates recent studies on the role and molecular mechanisms of ZFAS1 in malignancies and summarizes its clinical significance. By summarizing the role of ZFAS1 in cancer, we aim to highlight its potential as an anti-cancer biomarker and therapeutic target.
Collapse
Affiliation(s)
- Xin Liu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Zhong Ma
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Xianxu Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Shicheng Li
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Jiangdong An
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| | - Zhiqiang Luo
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
2
|
Luo B, Zhuang L, Huang J, Shi L, Zhang L, Zhu M, Lu Y, Zhu Q, Sun D, Wang H, Fang H. LncRNA ZFAS1 regulates ATIC transcription and promotes the proliferation and migration of hepatocellular carcinoma through the PI3K/AKT signaling pathway. J Cancer Res Clin Oncol 2024; 150:351. [PMID: 39001904 PMCID: PMC11246283 DOI: 10.1007/s00432-024-05877-1] [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: 02/27/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE Long noncoding RNAs (lncRNAs) exert a significant influence on various cancer-related processes through their intricate interactions with RNAs. Among these, lncRNA ZFAS1 has been implicated in oncogenic roles in multiple cancer types. Nevertheless, the intricate biological significance and underlying mechanism of ZFAS1 in the initiation and progression of hepatocellular carcinoma (HCC) remain largely unexplored. METHODS Analysis of The Cancer Genome Atlas Program (TCGA) database revealed a notable upregulation of lncRNA ZFAS1 in HCC tissues. To explore its function, we investigated colony formation and performed CCK-8 assays to gauge cellular proliferation and wound healing, Transwell assays to assess cellular migration, and an in vivo study employing a nude mouse model to scrutinize tumor growth and metastasis. Luciferase reporter assay was used to confirm the implicated interactions. Rescue experiments were conducted to unravel the plausible mechanism underlying the activation of the PI3K/AKT pathway by lncRNAs ZFAS1 and ATIC. RESULTS ZFAS1 and ATIC were significantly upregulated in the HCC tissues and cells. ZFAS1 knockdown inhibited cell proliferation and migration. We observed a direct interaction between the lncRNA ZFAS1 and ATIC. ATIC knockdown also suppressed cell proliferation and migration. SC79, an activator of AKT, partially restores the effects of lncRNA ZFAS1/ATIC knockdown on cell proliferation and migration. Knockdown of lncRNA ZFAS1/ATIC inhibited tumor growth and lung metastasis in vivo. CONCLUSION Overall, lncRNA ZFAS1 regulates ATIC transcription and contributes to the growth and migration of HCC cells through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Baoyang Luo
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, 185th Juqian Street, Changzhou, Jiangsu, 213003, China
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Lin Zhuang
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, 185th Juqian Street, Changzhou, Jiangsu, 213003, China
- Department of General Surgery, Wujin Affiliated Hospital of Jiangsu University and The Wujin Clinical college of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
| | - Ju Huang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, 226001, China
| | - Longqing Shi
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, 185th Juqian Street, Changzhou, Jiangsu, 213003, China
| | - Li Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, 185th Juqian Street, Changzhou, Jiangsu, 213003, China
| | - Maoqun Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, 185th Juqian Street, Changzhou, Jiangsu, 213003, China
| | - Yunjie Lu
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, 185th Juqian Street, Changzhou, Jiangsu, 213003, China
| | - Qiang Zhu
- Department of General Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Donglin Sun
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, 185th Juqian Street, Changzhou, Jiangsu, 213003, China.
| | - Hao Wang
- Department of General Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| | - Haisheng Fang
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
3
|
Mehrab Mohseni M, Zamani H, Momeni M, Shirvani-Farsani Z. An update on the molecular mechanisms of ZFAS1 as a prognostic, diagnostic, or therapeutic biomarker in cancers. Discov Oncol 2024; 15:219. [PMID: 38856786 PMCID: PMC11164845 DOI: 10.1007/s12672-024-01078-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024] Open
Abstract
Zinc finger antisense 1 (ZFAS1), a newly discovered long noncoding RNA, is expressed in various tissues and organs and has been introduced an oncogenic gene in human malignancies. In various cancers, ZFAS1 regulates apoptosis, cell proliferation, the cell cycle, migration, translation, rRNA processing, and spliceosomal snRNP assembly; targets signaling cascades; and interacts with transcription factors via binding to key proteins and miRNAs, with conflicting findings on its effect on these processes. ZFAS1 is elevated in different types of cancer, like colorectal, colon, osteosarcoma, and gastric cancer. Considering the ZFAS1 expression pattern, it also has the potential to be a diagnostic or prognostic marker in various cancers. The current review discusses the mode of action of ZFAS1 in various human cancers and its regulation function related to chemoresistance comprehensively, as well as the potential role of ZFAS1 as an effective and noninvasive cancer-specific biomarker in tumor diagnosis, prognosis, and treatment. We expected that the current review could fill the current scientific gaps in the ZFAS1-related cancer causative mechanisms and improve available biomarkers.
Collapse
Affiliation(s)
- Mahdieh Mehrab Mohseni
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, IR, Iran
| | - Hedyeh Zamani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, IR, Iran
| | - Mina Momeni
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, IR, Iran
| | - Zeinab Shirvani-Farsani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, IR, Iran.
| |
Collapse
|
4
|
Wang X, Nie P, Zhu D. LncRNA HOXA10-AS Activated by E2F1 Facilitates Proliferation and Migration of Nasopharyngeal Carcinoma Cells Through Sponging miR-582-3p to Upregulate RAB31. Am J Rhinol Allergy 2022; 36:348-359. [PMID: 35072529 DOI: 10.1177/19458924211064400] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a kind of head and neck cancer with a characteristic regional distribution. Increasing evidence has illustrated that long noncoding RNAs (lncRNAs) exert the regulatory function in tumor development. Nevertheless, the specific functions of lncRNA HOXA10 antisense RNA (HOXA10-AS) in NPC remain to be clarified. In this research, quantitative reverse transcription polymerase chain reaction detected HOXA10-AS expression in NPC cells. Cell counting kit-8, colony formation, and transwell assays were utilized to measure the proliferation and migration of NPC cells. Moreover, mechanism assays detected the interaction of different genes. Briefly, HOXA10-AS was highly expressed in NPC cells. HOXA10-AS down-regulation restrained NPC cell proliferation and migration. Further, HOXA10-AS could bind to miR-582-3p by acting as a competing endogenous RNA. Besides, Ras-related protein Rab-31 (RAB31) was proven as the target gene of miR-582-3p. Additionally, E2F transcription factor 1 (E2F1) acted as a transcription factor to activate HOXA10-AS expression. In the final rescue assays, we observed that the effect of HOXA10-AS depletion on NPC cell growth could be fully reversed by RAB31 overexpression or miR-582-3p inhibition. In short, our research proved that HOXA10-AS activated by E2F1 facilitated proliferation and migration of NPC cells through sponging miR-582-3p to upregulate RAB31.
Collapse
Affiliation(s)
- Xinyan Wang
- Jinan Fourth People's Hospital, Jinan, China
| | - Peilan Nie
- Jinan Fourth People's Hospital, Jinan, China
| | - Dongmei Zhu
- Jinan Fourth People's Hospital, Jinan, China
| |
Collapse
|
5
|
Wang G, Zhou Y, Zhong T, Song A, Xue Q. The role of blood lnc-ZFAS1 in acute ischemic stroke: correlation with neurological impairment, inflammation, and survival profiles. J Clin Lab Anal 2021; 36:e24219. [PMID: 34970793 PMCID: PMC8841186 DOI: 10.1002/jcla.24219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
Background Long non‐coding RNA zinc finger antisense 1 (lnc‐ZFAS1) has been reported to inhibit neuronal damage in acute ischemic stroke (AIS). However, the role of lnc‐ZFAS1 in AIS patients remains unclear. Therefore, we assessed the relationship of lnc‐ZFAS1 with neurological impairment, inflammation, and prognosis in AIS patients. Methods Totally, 241 AIS patients and 120 controls were enrolled. lnc‐ZFAS1 in peripheral blood mononuclear cells was evaluated using reverse transcription‐quantitative polymerase chain reaction. Besides, a 3‐year follow‐up was conducted to assess recurrence‐free survival (RFS) and overall survival (OS) in AIS patients. Results lnc‐ZFAS1 was reduced in AIS patients compared to that in controls (Z = −10.693, p < 0.001). In AIS patients, lnc‐ZFAS1 was negatively correlated with National Institutes of Health Stroke Scale score (rs = −0.335, p < 0.001), C‐reactive protein (rs = −0.284, p < 0.001), tumor necrosis factor‐alpha (rs = −0.293, p < 0.001), interleukin‐1β (rs = −0.149, p = 0.021), and interleukin‐6 (rs = −0.161, p = 0.012), but not underlying diseases (all p > 0.05). Besides, lnc‐ZFAS1 was divided into high and low levels based on the median expression in AIS patients. Indeed, high lnc‐ZFAS1 predicted better RFS (χ2 = 6.222, p = 0.013); the 1‐year, 2‐year, and 3‐year RFS rates were 94.2%, 88.3%, and 85.5%, respectively, in patients with high lnc‐ZFAS1, then 87.5%, 79.2%, and 71.6%, respectively, in those with low lnc‐ZFAS1. However, lnc‐ZFAS1 was not correlated with OS (χ2 = 1.404, p = 0.236); the 1‐year, 2‐year, and 3‐year OS rates were 98.3%, 95.8%, and 94.0%, respectively, in patients with high lnc‐ZFAS1, then 96.7%, 93.9%, and 89.6%, respectively, in those with low lnc‐ZFAS1. Conclusion Lower lnc‐ZFAS1 expression is connected with increased neurological impairment and inflammation as well as worse RFS in AIS patients.
Collapse
Affiliation(s)
- Gang Wang
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Ying Zhou
- Department of ICU, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Tingting Zhong
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Aixia Song
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Qian Xue
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| |
Collapse
|
6
|
Ma B, Cao L, Li Y. A novel 10-gene immune-related lncRNA signature model for the prognosis of colorectal cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:9743-9760. [PMID: 34814366 DOI: 10.3934/mbe.2021477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND The tumor immune microenvironment of colorectal cancer (CRC) affects tumor development, prognosis and immunotherapy strategies. Recently, immune-related lncRNA were shown to play vital roles in the tumor immune microenvironment. The objective of this study was to identify lncRNAs involved in the immune response, tumorigenesis and progression of CRC and to establish an immune-related lncRNA signature for predicting the prognosis of CRC. METHODS We used data retrieved from the cancer genome atlas (TCGA) dataset to construct a 10-gene immune-related lncRNA pair (IRLP) signature model using a method based on the ranking and comparison of paired gene expression in CRC. The clinical prognosis, immune checkpoints and lncRNA-protein networks were analyzed to evaluate the signature. RESULTS The signature was closely associated with overall survival of CRC patients (p < 0.001 in both of the training and validating cohorts) and the 3-year AUC values for the training and validating cohorts were 0.884 and 0.739, respectively. And, there were positive correlations between the signature and age (p = 0.048), clinical stage (p < 0.01), T stage (p < 0.01), N stage (p < 0.001) and M stage (p < 0.01). In addition, the signature model appeared to be highly relevant to some checkpoints, including CD160, TNFSF15, HHLA2, IDO2 and KIR3DL1. Further, molecular functional analysis and lncRNA-protein networks were applied to understand the molecular mechanisms underlying the carcinogenic effect and progression. CONCLUSION The 10-gene IRLP signature model is an independent prognostic factor for CRC patient and can be utilized for the development of immunotherapy.
Collapse
Affiliation(s)
- Bin Ma
- Department of Colorectal Surgery, Liaoning Cancer Hospital & Institute (Cancer Hospital of China Medical University), Shenyang, China
| | - Lianqun Cao
- Department of Colorectal Surgery, Liaoning Cancer Hospital & Institute (Cancer Hospital of China Medical University), Shenyang, China
| | - Yongmin Li
- Department of Colorectal Surgery, Liaoning Cancer Hospital & Institute (Cancer Hospital of China Medical University), Shenyang, China
| |
Collapse
|
7
|
Zhu H, Cheng Q, Cai H. lncRNA-ZFAS1 promotes the progression of endometrial carcinoma by targeting miR-34b to regulate VEGFA expression. Open Med (Wars) 2021; 16:1472-1481. [PMID: 34703899 PMCID: PMC8491586 DOI: 10.1515/med-2021-0362] [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: 02/28/2021] [Revised: 07/29/2021] [Accepted: 08/22/2021] [Indexed: 11/15/2022] Open
Abstract
Zinc finger nuclear transcription factor, X-box binding 1-type containing 1 antisense RNA 1 (ZFAS1) functions as an oncogenic long noncoding RNA (lncRNA) to promote proliferation and metastasis of endometrial carcinoma cell; however, the underlying mechanism has not been fully understood. First, RT-qPCR analysis of endometrial carcinoma tissues and cells showed that ZFAS1 was enriched in endometrial carcinoma tissues and cells. miR-34b was reduced in endometrial carcinoma and suggested negative correlation with ZFAS1 in endometrial carcinoma. Second, functional assays demonstrated that siRNA-mediated silence of ZFAS1 suppressed endometrial carcinoma cell proliferation and metastasis. Third, ZFAS1 bind to miR-34b and negatively regulate expression of miR-34b in endometrial carcinoma cells. miR-34b also bind to and negatively regulate expression of vascular endothelial growth factor A (VEGFA) in endometrial carcinoma cells. Lastly, knockdown of miR-34b counteracted with the suppressive effects of ZFAS1 silence on endometrial carcinoma cell proliferation and metastasis. In conclusion, lncRNA ZFAS1 functioned as an oncogene to promote endometrial carcinoma cell proliferation and metastasis through miR-34b/VEGFA axis.
Collapse
Affiliation(s)
- Hongli Zhu
- Department of Gynecology and Obstetrics, Affiliated Hangzhou First People's Hospital of Zhejiang University, No. 1 Xueshi Road, Shangcheng District, Hangzhou City, Zhejiang Province, 310010, China
| | - Qihui Cheng
- Department of Gynecology and Obstetrics, Affiliated Hangzhou First People's Hospital of Zhejiang University, No. 1 Xueshi Road, Shangcheng District, Hangzhou City, Zhejiang Province, 310010, China
| | - Hong Cai
- Department of Gynecology and Obstetrics, Affiliated Hangzhou First People's Hospital of Zhejiang University, No. 1 Xueshi Road, Shangcheng District, Hangzhou City, Zhejiang Province, 310010, China
| |
Collapse
|
8
|
Ghafouri-Fard S, Kamali MJ, Abak A, Shoorei H, Taheri M. LncRNA ZFAS1: Role in tumorigenesis and other diseases. Biomed Pharmacother 2021; 142:111999. [PMID: 34385106 DOI: 10.1016/j.biopha.2021.111999] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/23/2021] [Accepted: 08/01/2021] [Indexed: 12/28/2022] Open
Abstract
Residing on chromosome 20q13.13, Zinc Finger NFX1-Type Containing 1 (ZNFX1) antisense RNA 1 (ZFAS1) is a transcript which has been primarily recognized as a modulator of differentiation of alveolar and epithelial cell in the mammary gland. This long non-coding RNA (lncRNA) partakes in the molecular cascades leading to several non-neoplastic conditions such as osteoarthritis, epilepsy, rheumatoid arthritis, atherosclerosis, pulmonary fibrosis, myocardial infarction, and cardiac dysfunction. More importantly, ZFAS1 is considered as an oncogene in almost all types of cancers. Using expression amounts of ZFAS1, it is possible to forecast the clinical outcome of patients with different neoplasms such as colorectal cancer, gastric cancer, cholangiocarcinoma, hepatoblastoma, and other types of cancer. We describe the role of ZFAS1 in the development of neoplastic and non-neoplastic disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Atefe Abak
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Wang R, Wang X, Zhang J, Liu Y. LINC00942 Promotes Tumor Proliferation and Metastasis in Lung Adenocarcinoma via FZD1 Upregulation. Technol Cancer Res Treat 2021; 20:1533033820977526. [PMID: 34253104 PMCID: PMC8280845 DOI: 10.1177/1533033820977526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have been reported to play important roles in the progression of human cancers. Herein, bioinformatic analysis identified that LINC00942 was a highly overexpressed lncRNA in lung adenocarcinoma (LUAD). The present study aimed to explore the roles and possible molecular mechanisms of LINC00942 in LUAD. METHODS First, on the basis of TCGA database, the expression and prognosis of LINC00942 were analyzed in LUAD tissues. Then, si-LINC00942 was transfected into A549 and H1299 cells to knockdown the expression of LINC00942. Cell viability was detected by MTT assay. Flow cytometry was used to analyze cell apoptosis. The expressions of PCNA, Bax, Bcl-2, and wnt/β-catenin pathway proteins were detected by western blotting. Dual-luciferase reporter assay was used to evaluate the regulatory relationship between LINC00942 and miR-5006-5p, or miR-5006-5p and FZD1. RESULTS We discovered that LINC00942 was up-regulated in LUAD tissues compared with adjacent tissues. Besides, we found the increased LINC00942 expression was associated with poor survival. In addition, silencing of LINC00942 suppressed the proliferation, migration, invasion and facilitated the apoptosis of A549 and H1299 cells. Moreover, silencing of LINC00942 repressed the expression of PCNA, Bcl-2, and enhanced Bax expression in A549 and H1299 cells. Mechanically, LINC00942 exerted its effects via enhancing Wnt signaling. LINC00942 functioned as competing endogenous RNA (ceRNA) by binding to miR-5006-5p, upregulating the expression of FZD1, which was a direct target of miR-5006-5p. CONCLUSION Our findings indicated that LINC00942/miR-5006-5p/FZD1 axis played important roles in LUAD growth through enhancing Wnt signaling. LINC00942/miR-5006-5p/FZD1 axis might serve as a potential biomarker and therapeutic target for LUAD treatment.
Collapse
Affiliation(s)
- Ronghua Wang
- Department of Outpatient, Dongying People's Hospital, Dongying, Shandong, People's Republic of China
| | - Xiuyun Wang
- Department of Oncology, Dongying People's Hospital, Dongying, Shandong, People's Republic of China
| | - Jingtao Zhang
- Department of Respiratory Medicine, Dongying District People's Hospital of Dongying city, Dongying, Shandong, People's Republic of China
| | - Yanpei Liu
- Department of Outpatient, Dongying People's Hospital, Dongying, Shandong, People's Republic of China
| |
Collapse
|
10
|
Wang H, Hu H, Ma J, Jiang Y, Cheng R. lncRNA ZFAS1 promotes the ox-LDL induced proliferation, invasion and migration of vascular smooth muscle cells. Exp Ther Med 2021; 22:835. [PMID: 34149881 PMCID: PMC8200810 DOI: 10.3892/etm.2021.10267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/18/2021] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory vascular disease. The dysfunction of vascular smooth muscle cells (VSMCs) induced by oxidized low-density lipoprotein (ox-LDL) contributes to the formation of atherosclerotic lesions. Additionally, upregulation of the long non-coding RNA zinc finger antisense 1 (ZFAS1) was observed in the plaques of patients with atherosclerosis. The aim of the present study was to explore the functional role of ZFAS1 in atherosclerosis progression. Reverse transcription-quantitative PCR was performed to analyze ZFAS1 mRNA expression, and western blotting was performed to determine the protein expression levels of Ki67, proliferating cell nuclear antigen (PCNA), matrix metallopeptidase (MMP)2 and MMP9. The Cell Counting Kit-8 assay was used to test cell viability. Finally, wound healing and Transwell chamber assays were performed to evaluate cell migration and invasion, respectively. The current findings demonstrated that ZFAS1 expression was upregulated by ox-LDL stimulation in VSMCs. Moreover, ZFAS1 overexpression promoted the ox-LDL-induced proliferation, migration and invasion of VSMCs, and upregulated the expression levels of proteins associated with cellular proliferation (Ki67 and PCNA), migration and invasion (MMP2 and 9). By contrast, ZFAS1-knockdown inhibited the proliferation, migration and invasion of VSMCs, and suppressed cell proliferation-, migration- and invasion-associated protein expression. In conclusion, ZFAS1 promoted the ox-LDL-induced proliferation, invasion and migration of VSMCs. Thus, ZFAS1 may represent a novel biomarker for dysfunction of VSMCs in the pathological condition of atherosclerosis.
Collapse
Affiliation(s)
- Hao Wang
- Department of Clinical Laboratory, Linhai Traditional Chinese Medicine Hospital Medical Community, Linhai, Zhejiang 317000, P.R. China
| | - Huajie Hu
- Department of Clinical Laboratory, Linhai Traditional Chinese Medicine Hospital Medical Community, Linhai, Zhejiang 317000, P.R. China
| | - Junjie Ma
- Department of Clinical Laboratory, Linhai Traditional Chinese Medicine Hospital Medical Community, Linhai, Zhejiang 317000, P.R. China
| | - Yafeng Jiang
- Department of Medical Genetics and Prenatal Diagnosis, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu 222000, P.R. China
| | - Ruifei Cheng
- Department of Clinical Laboratory, Lianyungang Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Lianyungang, Jiangsu 222004, P.R. China
| |
Collapse
|
11
|
Tang Y, He X. Long non-coding RNAs in nasopharyngeal carcinoma: biological functions and clinical applications. Mol Cell Biochem 2021; 476:3537-3550. [PMID: 33999333 DOI: 10.1007/s11010-021-04176-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common head and neck malignancies. It has obvious ethnic and regional specificity. Long non-coding RNAs (LncRNAs) are a class of non-protein coding RNA molecules. Emerging research shows that lncRNAs play a key role in tumor development, prognosis, and treatment. With the deepening of sequence analysis, a large number of functional LncRNAs have been found in NPC, which interact with coding genes, miRNAs, and proteins to form a complex regulatory network. However, the specific role and mechanism of abnormally expressed lncRNAs in the pathogenesis of NPC is not fully understood. This article briefly introduced the concept, classification, and functional mechanism of lncRNAs and reviewed their biological functions and their clinical applications in NPC. Specifically, we described lncRNAs related to the occurrence, growth, invasion, metastasis, angiogenesis, and cancer stem cells of NPC; discussed lncRNAs related to Epstein-Barr virus infection; and summarized the role of lncRNAs in NPC treatment resistance. We have also sorted out lncRNAs related to Chinese medicine treatment. We believe that with the deepening of lncRNAs research, tumor-specific lncRNAs may become a new target for the treatment and a biomarker for predicting prognosis.
Collapse
Affiliation(s)
- Yao Tang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology (2016TP1015), Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, 421001, Hunan Province, China
| | - Xiusheng He
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology (2016TP1015), Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|
12
|
Peng J, Liu F, Zheng H, Wu Q, Liu S. IncRNA ZFAS1 contributes to the radioresistance of nasopharyngeal carcinoma cells by sponging hsa-miR-7-5p to upregulate ENO2. Cell Cycle 2020; 20:126-141. [PMID: 33342344 DOI: 10.1080/15384101.2020.1864128] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Previous research revealed that lncRNA ZFAS1 could promote nasopharyngeal carcinoma (NPC) by inhibiting its downstream target axis. However, the association between ZFAS1 and radioresistant NPC cells is unclear. This study aimed to explore the roles of ZFAS1 in the radioresistance of NPC. Bioinformatics analysis was conducted to identify the significant factors (ENO2 and miR-7-5p) that contributed to the radioresistance of NPC cells. After performing qRT-PCR analysis, we found that the expression of ZFAS1 and ENO2 was upregulated in NPC cells but that the miR-7-5p expression was downregulated in the same samples. Apart from that, we noticed that ZFAS1 inhibition enhanced the sensitivity of NPC cells to radiation therapy by repressing cell proliferation and promoting cell apoptosis. Subsequently, we found that ZFAS1 could sponge miR-7-5p to upregulate ENO2, which was the target of miR-7-5p. Experimental results also indicated that the suppression of miR-7-5p inhibited the sensitivity of NPC cells to radiation therapy, thereby suppressing ENO2 expression. Overall, our findings suggested that ZFAS1 contributed to the radioresistance of NPC cells by regulating the miR-7-5p/ENO2 axis and that ZFAS1 might be a potential therapeutic target for addressing the radioresistance of NPC cells.
Collapse
Affiliation(s)
- Jiaojiao Peng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University , Sichuan, China
| | - Feng Liu
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University , Sichuan, China
| | - Hong Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University , Sichuan, China
| | - Qi Wu
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University , Sichuan, China
| | - Shixi Liu
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University , Sichuan, China
| |
Collapse
|
13
|
Sun Y, Gao X, Li P, Song L, Shi L. LncRNA ZFAS1, as a poor prognostic indicator, promotes cell proliferation and epithelial-mesenchymal transition in endometrial carcinoma. Per Med 2020; 18:43-53. [PMID: 33151128 DOI: 10.2217/pme-2020-0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: Long noncoding RNA Zinc finger nuclear transcription factor, X-box binding 1-type containing 1 antisense RNA 1 (ZFAS1) has been reported to be an oncogene in various tumors. However, the role of ZFAS1 in endometrial carcinoma (EC) are not fully determined. Methods & results: Here, we found ZFAS1 expression was significantly upregulated in EC patients, which was significantly associated with International Federation of Gynecology and Obstetrics stage, histological grade, myometrial invasion and poor prognosis. The loss-of-function assays showed that knockdown of ZFAS1 significantly suppressed the proliferation, G1/S transition, migration and invasion in EC cells. Moreover, knockdown of ZFAS1 obviously downregulated the expression of CDK4, Cyclin D1 and N-cadherin, but upregulated E-cadherin expression. Conclusion: Collectively, these results suggest that ZFAS1 might be used as potential therapeutic targets for EC treatment.
Collapse
Affiliation(s)
- Yanan Sun
- Department of Gynecology, The Second Affiliated Hospital Harbin Medical University, Harbin, Heilongjiang, PR China.,Department of Gynecology & Obstetrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang, PR China
| | - Xuan Gao
- Department of Gynecology & Obstetrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang, PR China
| | - Peiling Li
- Department of Gynecology, The Second Affiliated Hospital Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Ling Song
- Department of Gynecology & Obstetrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang, PR China
| | - Lei Shi
- Department of Gynecology & Obstetrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang, PR China
| |
Collapse
|
14
|
You H, Wang S, Yu S. KIF9-AS1 promotes nasopharyngeal carcinoma progression by suppressing miR-16. Oncol Lett 2020; 20:241. [PMID: 32973955 PMCID: PMC7509506 DOI: 10.3892/ol.2020.12104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/19/2020] [Indexed: 01/07/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been reported to serve a crucial role in the progression of nasopharyngeal carcinoma (NPC); however, the underlying molecular mechanisms of lncRNA KIF9-AS1 in the tumorigenesis of NPC remains poorly understood. Reverse transcription-quantitative PCR was used to analyze the expression levels of KIF9-AS1 and microRNA (miR)-16, and Cell Counting Kit-8, wound healing and Transwell assays were used to determine the cell viability, invasion and migration, respectively, of NPC cells. In addition, a dual-luciferase reporter assay was used to analyze the direct interaction between KIF9-AS1 and miR-16. NPC stage was classified according to the seventh edition of the AJCC staging system. The results revealed that KIF9-AS1 expression levels were upregulated in NPC tissues and cell lines. In addition, miR-16 was demonstrated to directly interact with KIF9-AS1 and inhibit KIF9-AS1 expression levels, whereas the miR-16 inhibitor rescued the effects of the KIF9-AS1-knockdown in NPC cells. Furthermore, the expression levels of KIF9-AS1 were upregulated, while those of miR-16 were downregulated in NPC tissues. Notably, the expression levels of KIF9-AS1 were observed to be significantly more upregulated in advanced tumors (III–IV vs. I–II) and patients with high KIF9-AS1 expression levels exhibited a worse prognosis. In conclusion, the findings of the present study suggested that KIF9-AS1 may promote the progression of NPC by targeting miR-16, thus KIF9-AS1 may be a novel molecular target for NPC therapy.
Collapse
Affiliation(s)
- Huizeng You
- Department of Otolaryngology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261041, P.R. China
| | - Shuyong Wang
- Department of Otolaryngology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261041, P.R. China
| | - Sa Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhuji People's Hospital of Zhejiang Province, Zhuji, Zhejiang 311800, P.R. China
| |
Collapse
|
15
|
Yang X, Lin F, Gao F. Up-regulated long non-coding RNA ILF3-AS1 indicates poor prognosis of nasopharyngeal carcinoma and promoted cell metastasis. Int J Biol Markers 2020; 35:61-70. [PMID: 32962500 DOI: 10.1177/1724600820955199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have been confirmed to participate in the regulation of nasopharyngeal carcinoma. Here, we endeavored to explore the character of lncRNA ILF3-AS1 in the nasopharyngeal carcinoma and its function. METHODS A total of 68 nasopharyngeal carcinoma tissues and adjacent normal nasopharyngeal tissues were collected. Expressions of lncRNA ILF3-AS1 in these tissues were detected using quantitative real-time polymerase chain reaction (qRT-PCR). The relationship between the expression level of lncRNA ILF3-AS1 and clinical pathological characteristics was analyzed. Inhibition of lncRNA ILF3-AS1 was done using small interference RNA. RESULTS lncRNA ILF3-AS1 expression was significantly up-regulated in the 68 nasopharyngeal carcinoma tissue samples compared to their adjacent normal tissue samples. Increased lncRNA ILF3-AS1 level was related to the advanced tumor node metastasis stage and the metastasis of nasopharyngeal carcinoma. Also, increased lncRNA ILF3-AS1 indicated poor prognosis of nasopharyngeal carcinoma patients. Inhibition of lncRNA ILF3-AS1 reduced proliferation, invasion and migration of nasopharyngeal carcinoma cells. MicroRNA-320a (miR-320a) was determined as a direct target for lncRNA ILF3-AS1 in nasopharyngeal carcinoma. Furthermore, lncRNA ILF3-AS1 could sponge miR-320a to promote BMI1 expression. The expression of BMI1 was significantly inhibited by the down-regulation of lncRNA ILF3-AS1. CONCLUSIONS For the first time, we demonstrated that lncRNA ILF3-AS1 was markedly over-expressed in nasopharyngeal carcinoma tissues and cells. Elevated lncRNA ILF3-AS1 expression was correlated with severe cancer stage and poor prognosis. lncRNA ILF3-AS1 could promote proliferation, invasion, and migration of cells, which might indicate a novel target site for the future diagnosis and therapy of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Xuewen Yang
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Lin
- Hainan Medical University, Hainan, China
| | - Feng Gao
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
16
|
Hu F, Shao L, Zhang J, Zhang H, Wen A, Zhang P. Knockdown of ZFAS1 Inhibits Hippocampal Neurons Apoptosis and Autophagy by Activating the PI3K/AKT Pathway via Up-regulating miR-421 in Epilepsy. Neurochem Res 2020; 45:2433-2441. [DOI: 10.1007/s11064-020-03103-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/02/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022]
|
17
|
Fan G, Jiao J, Shen F, Chu F. Upregulation of lncRNA ZFAS1 promotes lung adenocarcinoma progression by sponging miR-1271-5p and upregulating FRS2. Thorac Cancer 2020; 11:2178-2187. [PMID: 32515146 PMCID: PMC7396366 DOI: 10.1111/1759-7714.13525] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/10/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022] Open
Abstract
Background Nowadays, the important roles of long non‐coding RNAs (LncRNAs) in lung adenocarcinoma (LAD) is being increasingly recognized. The purpose of this study was to explore the regulatory mechanism of lncRNA ZFAS1 in LAD. Methods The expression and function of lncRNA ZFAS1 were assessed by RT‐qPCR, CCK‐8, transwell and dual luciferase reporter assays. Results Upregulation of lncRNA ZFAS1 was found in LAD tissues and cells. Knockdown of lncRNA ZFAS1 restrained cell proliferation, migration and invasion in LAD cells. In addition, we determined that lncRNA ZFAS1 could directly bind to miR‐1271‐5p. MiR‐1271‐5p functioned as a tumor suppressor in LAD, and lncRNA ZFAS1 promoted LAD development by downregulating miR‐1271‐5p. Furthermore, FRS2 was a direct target of miR‐1271‐5p. FRS2 promoted progression of LAD by mediating lncRNA ZFAS1/miR‐1271‐5p axis. Conclusions LncRNA ZFAS1 promotes cell proliferation, migration and invasion in LAD by downregulating miR‐1271‐5p or upregulating FRS2.
Collapse
Affiliation(s)
- Gang Fan
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jin Jiao
- Department of Clinical Laboratory, Shandong Maternal and Child Health Care Hospital, Jinan, China
| | - Feng Shen
- Department of Clinical Laboratory, Shandong Maternal and Child Health Care Hospital, Jinan, China
| | - Fulu Chu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
18
|
Jiang N, Dai Q, Su X, Fu J, Feng X, Peng J. Role of PI3K/AKT pathway in cancer: the framework of malignant behavior. Mol Biol Rep 2020; 47:4587-4629. [PMID: 32333246 PMCID: PMC7295848 DOI: 10.1007/s11033-020-05435-1] [Citation(s) in RCA: 322] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/03/2020] [Indexed: 12/12/2022]
Abstract
Given that the PI3K/AKT pathway has manifested its compelling influence on multiple cellular process, we further review the roles of hyperactivation of PI3K/AKT pathway in various human cancers. We state the abnormalities of PI3K/AKT pathway in different cancers, which are closely related with tumorigenesis, proliferation, growth, apoptosis, invasion, metastasis, epithelial-mesenchymal transition, stem-like phenotype, immune microenvironment and drug resistance of cancer cells. In addition, we investigated the current clinical trials of inhibitors against PI3K/AKT pathway in cancers and found that the clinical efficacy of these inhibitors as monotherapy has so far been limited despite of the promising preclinical activity, which means combinations of targeted therapy may achieve better efficacies in cancers. In short, we hope to feature PI3K/AKT pathway in cancers to the clinic and bring the new promising to patients for targeted therapies.
Collapse
Affiliation(s)
- Ningni Jiang
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Qijie Dai
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Xiaorui Su
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Jianjiang Fu
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Xuancheng Feng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Juan Peng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| |
Collapse
|