1
|
Chen X, Jiang R, Huang X, Chen L, Hu X, Wei Y. Long Noncoding RNA NKX2-1-AS1 Accelerates Non-Small Cell Lung Cancer Progression through the miR-589-5p/NME1 Axis. Cell Biochem Biophys 2024:10.1007/s12013-024-01472-w. [PMID: 39117986 DOI: 10.1007/s12013-024-01472-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Non-small cell lung cancer (NSCLC) is the most common malignancy worldwide, with a high death rate. Long noncoding RNA (LncRNA) NKX2-1 antisense RNA 1 (NKX2-1-AS1) has been reported to be an oncogene in lung tumorigenesis. However, the precise mechanism of NKX2-1-AS1 underlying NSCLC progression is blurry. The intention of our research was to probe the potential mechanism of NKX2-1-AS1 underlying NSCLC. NKX2-1-AS1 expression and relevant downstream gene expression were measured using RT-qPCR. Cell proliferation and apoptosis were determined by MTT assay, EdU assay along with flow cytometry analysis. Cell migratory and invasive abilities were inspected by transwell assay. Western blot and immunofluorescence staining were utilized to assess the levels of epithelial-mesenchymal transition (EMT)-related proteins. RNA pull-down together with luciferase reporter assays were performed to verify the interaction between NKX2-1-AS1 and its downstream RNAs. Xenograft tumor-bearing mouse models were built to analyze tumor growth in vivo. The results suggested that NKX2-1-AS1 was upregulated in NSCLC patient tissues and cell lines. NKX2-1-AS1 deficiency suppressed cell proliferation, migration, invasion and EMT while elevated apoptosis. NKX2-1-AS1 bound to miR-589-5p, and NME/NM23 nucleoside diphosphate kinase 1 (NME1) was targeted by miR-589-5p in NSCLC cells. Additionally, NKX2-1-AS1 accelerated the progression of NSCLC by regulating miR-589-5p/NME1 axis. NKX2-1-AS1 knockdown repressed tumor growth in vivo. In conclusion, NKX2-1-AS1 accelerated the NSCLC progression through interacting with miR-589-5p for NME1 upregulation, which may provide clues for NSCLC targeting therapy.
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Ruilai Jiang
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Xiaocheng Huang
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Ling Chen
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Xiaogang Hu
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Yanbin Wei
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China.
| |
Collapse
|
2
|
Yan Q, Wong W, Gong L, Yang J, Liang D, Chin KY, Dai S, Wang J. Roles of long non‑coding RNAs in esophageal cell squamous carcinoma (Review). Int J Mol Med 2024; 54:72. [PMID: 38963019 PMCID: PMC11232667 DOI: 10.3892/ijmm.2024.5396] [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: 02/24/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent and deadly malignancy of the digestive tract. Recent research has identified long non‑coding RNAs (lncRNAs) as crucial regulators in the pathogenesis of ESCC. These lncRNAs, typically exceeding 200 nucleotides, modulate gene expression through various mechanisms, including the competing endogenous RNA (ceRNA) pathway and RNA‑protein interactions. The current study reviews the multifaceted roles of lncRNAs in ESCC, highlighting their involvement in processes such as proliferation, migration, invasion, epithelial‑mesenchymal transition, cell cycle progression, resistance to radiotherapy and chemotherapy, glycolysis, apoptosis, angiogenesis, autophagy, tumor growth, metastasis and the maintenance of cancer stem cells. Specific lncRNAs like HLA complex P5, LINC00963 and non‑coding repressor of NFAT have been shown to enhance resistance to radio‑ and chemotherapy by modulating pathways such as AKT signaling and microRNA interaction, which promote cell survival and proliferation under therapeutic stress. Furthermore, lncRNAs like family with sequence similarity 83, member A antisense RNA 1, zinc finger NFX1‑type containing 1 antisense RNA 1 and taurine upregulated gene 1 are implicated in enhancing invasive and proliferative capabilities of ESCC cells through the ceRNA mechanism, while interactions with RNA‑binding proteins further influence cancer cell behavior. The comprehensive analysis underscores the potential of lncRNAs as biomarkers for prognosis and therapeutic targets in ESCC, suggesting avenues for future research focused on elucidating the detailed molecular mechanisms and clinical applications of lncRNAs in ESCC management.
Collapse
Affiliation(s)
- Qihang Yan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong 510060, P.R. China
| | - Wingshing Wong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Li Gong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jie Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Dachuan Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia
| | - Shuqin Dai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Junye Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
3
|
Wei Q, Liu G, Huang Z, Nian J, Huang L, Huang Y, Huang Z, Pu J. lncRNA PAARH impacts liver cancer cell proliferation by engaging miR‑6512‑3p to target LASP1. Oncol Lett 2024; 28:306. [PMID: 38774456 PMCID: PMC11106750 DOI: 10.3892/ol.2024.14439] [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: 10/03/2023] [Accepted: 03/18/2024] [Indexed: 05/24/2024] Open
Abstract
Long non-coding (lnc)RNAs serve a pivotal role as regulatory factors in carcinogenesis. The present study aimed to assess the involvement of the lncRNA progression and angiogenesis-associated RNA in hepatocellular carcinoma (PAARH) in liver cancer, along with the associated underlying mechanism. Through the use of reverse transcription-quantitative (RT-q)PCR, differences in the expression levels of PAARH in HepG2, HEP3B2.1.7, HCCLM3, Huh-7 and MHCC97-H liver cancer cell lines and THLE-2 epithelial cell lines were evaluated. The liver cancer cell line with the greatest, significantly different, level of expression relative to the normal liver cell line was selected for subsequent experiments. Using ENCORI database, the putative target genes of the microRNA (miR) miR-6512-3p were predicted. Cells were then transfected with lentiviruses carrying short-hairpin-PAARH to interfere with PAARH expression. Subsequently, HepG2 liver cancer cells were transfected with a miR-6512-3p mimic and an inhibitor, and the expression levels of miR-6512-3p and the LIM and SH3 domain protein 1 (LASP1) in cells were assessed using RT-qPCR analysis. Cell proliferation was subsequently evaluated using colony formation assays, and immunofluorescence and western blotting were used to assess the expression level of LASP1 in transfected cells. The binding interaction between miR-6512-3p and LASP1 was further evaluated using a dual-luciferase reporter gene assay. Liver cancer cells were found to exhibit higher expression levels of PAARH compared with normal liver cells. Following PAARH interference, the expression level of miR-6512-3p was significantly increased, whereas that of LASP1 was significantly decreased, resulting in a reduction in cell proliferation. In liver cancer cells, miR-6512-3p overexpression led to a significant reduction in the LASP1 level and reduced proliferation, whereas suppressing miR-6512-3p led to a significant increase in LASP1 levels and increased proliferation. Additionally, the inhibition of miR-6512-3p caused the states of low LASP1 expression and reduced cell proliferation to be reversed. LASP1, a recently identified target gene of miR-6512-3p, was demonstrated to be suppressed by miR-6512-3p overexpression, thereby inhibiting liver cancer cell proliferation. Taken together, the findings of the present study demonstrate that the lncRNA PAARH may enhance liver cancer cell proliferation by engaging miR-6512-3p to target LASP1.
Collapse
Affiliation(s)
- Qing Wei
- Graduate College, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Guoman Liu
- Graduate College, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zihua Huang
- Graduate College, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jiahui Nian
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Lizheng Huang
- Graduate College, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yanyan Huang
- Graduate College, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zheng Huang
- Graduate College, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jian Pu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| |
Collapse
|
4
|
Barbato A, Piscopo F, Salati M, Pollastro C, Evangelista L, Ferrante L, Limongello D, Brillante S, Iuliano A, Reggiani-Bonetti L, Salatiello M, Iaccarino A, Pisapia P, Malapelle U, Troncone G, Indrieri A, Dominici M, Franco B, Carotenuto P. A MiR181/Sirtuin1 regulatory circuit modulates drug response in biliary cancers. Clin Exp Med 2024; 24:74. [PMID: 38598008 PMCID: PMC11006774 DOI: 10.1007/s10238-024-01332-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024]
Abstract
Despite recent advances, biliary tract cancer (BTC) remains one of the most lethal tumor worldwide due to late diagnosis, limited therapeutic strategies and resistance to conventional therapies. In recent years, high-throughput technologies have enabled extensive genome, and transcriptome sequencing unveiling, among others, the regulatory potential of microRNAs (miRNAs). Compelling evidence shown that miRNA are attractive therapeutic targets and promising candidates as biomarkers for various therapy-resistant tumors. The analysis of miRNA profile successfully identified miR-181c and -181d as significantly downregulated in BTC patients. Low miR-181c and -181d expression levels were correlated with worse prognosis and poor treatment efficacy. In fact, progression-free survival analysis indicated poor survival rates in miR-181c and -181d low expressing patients. The expression profile of miR-181c and -181d in BTC cell lines revealed that both miRNAs were dysregulated. Functional in vitro experiments in BTC cell lines showed that overexpression of miR-181c and -181d affected cell viability and increased sensitivity to chemotherapy compared to controls. In addition, by using bioinformatic tools we showed that the miR-181c/d functional role is determined by binding to their target SIRT1 (Sirtuin 1). Moreover, BTC patients expressing high levels of miR-181 and low SIRT1 shown an improved survival and treatment response. An integrative network analysis demonstrated that, miR-181/SIRT1 circuit had a regulatory effect on several important metabolic tumor-related processes. Our study demonstrated that miR-181c and -181d act as tumor suppressor miRNA in BTC, suggesting the potential use as therapeutic strategy in resistant cancers and as predictive biomarker in the precision medicine of BTC.
Collapse
Affiliation(s)
- Anna Barbato
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy
| | - Fabiola Piscopo
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy
| | - Massimiliano Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41125, Modena, Italy
| | - Carla Pollastro
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy
| | - Lorenzo Evangelista
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Luigi Ferrante
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Davide Limongello
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Simona Brillante
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- IRGB, Institute for Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Antonella Iuliano
- Department of Mathematics, Computer Science and Economics (DIMIE), University of Basilicata, 85100, Potenza, Italy
| | - Luca Reggiani-Bonetti
- Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, 41125, Modena, Italy
| | - Maria Salatiello
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Antonino Iaccarino
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Alessia Indrieri
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- IRGB, Institute for Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41125, Modena, Italy
| | - Brunella Franco
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, 80078, Naples, Italy
| | - Pietro Carotenuto
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy.
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy.
| |
Collapse
|
5
|
Liu S, Li W, Liang L, Zhou Y, Li Y. The regulatory relationship between transcription factor STAT3 and noncoding RNA. Cell Mol Biol Lett 2024; 29:4. [PMID: 38172648 PMCID: PMC10763091 DOI: 10.1186/s11658-023-00521-1] [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: 09/30/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3), as a key node in numerous carcinogenic signaling pathways, is activated in various tumor tissues and plays important roles in tumor formation, metastasis, and drug resistance. STAT3 is considered a potential subtarget for tumor therapy. Noncoding RNA (ncRNA) is a special type of RNA transcript. Transforming from "junk" transcripts into key molecules involved in cell apoptosis, growth, and functional regulation, ncRNA has been proven to be closely related to various epithelial-mesenchymal transition and drug resistance processes in tumor cells over the past few decades. Research on the relationship between transcription factor STAT3 and ncRNAs has attracted increased attention. To date, existing reviews have mainly focused on the regulation by ncRNAs on the transcription factor STAT3; there has been no review of the regulation by STAT3 on ncRNAs. However, understanding the regulation of ncRNAs by STAT3 and its mechanism is important to comprehensively understand the mutual regulatory relationship between STAT3 and ncRNAs. Therefore, in this review, we summarize the regulation by transcription factor STAT3 on long noncoding RNA, microRNA, and circular RNA and its possible mechanisms. In addition, we provide an update on research progress on the regulation of STAT3 by ncRNAs. This will provide a new perspective to comprehensively understand the regulatory relationship between transcription factor STAT3 and ncRNAs, as well as targeting STAT3 or ncRNAs to treat diseases such as tumors.
Collapse
Affiliation(s)
- Siyi Liu
- Department of Nuclear Medicine, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Wentao Li
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Lin Liang
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China.
| | - Yanling Li
- Department of Nuclear Medicine, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China.
| |
Collapse
|
6
|
Huang T, Wu Z, Zhu S. The roles and mechanisms of the lncRNA-miRNA axis in the progression of esophageal cancer: a narrative review. J Thorac Dis 2022; 14:4545-4559. [PMID: 36524088 PMCID: PMC9745524 DOI: 10.21037/jtd-22-1449] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/08/2022] [Indexed: 12/08/2023]
Abstract
BACKGROUND AND OBJECTIVE Esophageal cancer is one of the most common malignant digestive tract tumors. Despite various treatment methods, the prognosis of patients remains unsatisfactory, largely due to an insufficient understanding of the mechanisms involved in the pathogenesis and progression of esophageal cancer. More than 98% of the nucleotide sequences in the human genome do not encode proteins, and their transcription products are noncoding RNAs (ncRNAs), mainly long noncoding RNAs (lncRNAs) and microRNAs (miRNAs). Experiments have shown that lncRNAs and miRNAs play crucial roles in the occurrence and progression of various human malignancies. These ncRNAs influence the progression of esophageal cancer through an intricate regulatory network. We herein summarized the roles and mechanisms of the lncRNA-miRNA axis in esophageal cancer cell proliferation, apoptosis, epithelial-mesenchymal transition (EMT), invasion and metastasis, drug resistance, radiotherapy resistance, and angiogenesis. This review provides a rationale for anticancer therapy that targets the lncRNA-miRNA axis in esophageal cancer. METHODS Related articles published in the PubMed database between 05/30/2008 to 09/10/2022 were identified using the following terms: "lncRNA AND miRNA AND esophageal cancer", "lncRNA AND miRNA AND cell proliferation", "lncRNA AND miRNA AND apoptosis", "lncRNA AND miRNA AND EMT", "lncRNA AND miRNA AND invasion and metastasis", "lncRNA AND miRNA AND drug resistance", and "lncRNA AND miRNA AND radiotherapy resistance". Published articles written in English available to readers were considered. KEY CONTENT AND FINDINGS We summarized the roles of the lncRNA-miRNA axis in the progression of esophageal cancer, including cell proliferation, apoptosis, EMT, invasion and metastasis, drug resistance, radio resistance, and other progressions, and determined that the lncRNA-miRNA axis may serve as a potential clinical treatment target for esophageal cancer. CONCLUSIONS The lncRNA-miRNA axis is closely related to the progression of esophageal cancer and may act as a potential biological target for the clinical treatment of patients with esophageal cancer.
Collapse
Affiliation(s)
- Tao Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, China
| | - Zhihao Wu
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, China
| | - Shaojin Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| |
Collapse
|