1
|
Zarandi PK, Ghiasi M, Heiat M. The role and function of lncRNA in ageing-associated liver diseases. RNA Biol 2025; 22:1-8. [PMID: 39697114 PMCID: PMC11660375 DOI: 10.1080/15476286.2024.2440678] [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] [Revised: 10/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Liver diseases are a significant global health issue, characterized by elevated levels of disorder and death. The substantial impact of ageing on liver diseases and their prognosis is evident. Multiple processes are involved in the ageing process, which ultimately leads to functional deterioration of this organ. The process of liver ageing not only renders the liver more susceptible to diseases but also compromises the integrity of other organs due to the liver's critical function in metabolism regulation. A growing body of research suggests that long non-coding RNAs (lncRNAs) play a significant role in the majority of pathophysiological pathways. They regulate gene expression through a variety of interactions with microRNAs (miRNAs), messenger RNAs (mRNAs), DNA, or proteins. LncRNAs exert a major influence on the progression of age-related liver diseases through the regulation of cell proliferation, necrosis, apoptosis, senescence, and metabolic reprogramming. A concise overview of the current understanding of lncRNAs and their potential impact on the development of age-related liver diseases will be provided in this mini-review.
Collapse
Affiliation(s)
- Peyman Kheirandish Zarandi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohsen Ghiasi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Evangelista JE, Ali-Nasser T, Malek LE, Xie Z, Marino GB, Bester AC, Ma'ayan A. lncRNAlyzr: Enrichment Analysis for lncRNA Sets. J Mol Biol 2025; 437:168938. [PMID: 40133794 PMCID: PMC12145269 DOI: 10.1016/j.jmb.2025.168938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 03/27/2025]
Abstract
lncRNAs make up a large portion of the human genome affecting many biological processes in normal physiology and diseases. However, human lncRNAs are understudied compared to protein-coding genes. While there are many tools for performing gene set enrichment analysis for coding genes, few tools exist for lncRNA enrichment analysis. lncRNAlyzr is a webserver application designed for lncRNAs enrichment analysis. lncRNAlyzr has a database containing 33 lncRNA set libraries created by computing correlations between lncRNAs and annotated coding gene sets. After users submit a set of lncRNAs to lncRNAlyzr, the enrichment analysis results are visualized as ball-and-stick subnetworks where nodes are lncRNAs connected to enrichment terms from across selected lncRNA set libraries. To demonstrate lncRNAlyzr, it was used to analyze the effects of knocking down the lncRNA CYTOR in K562 cells. Overall, lncRNAlyzr is an enrichment analysis tool for lncRNAs aiming to further our understanding of lncRNAs functional modules. lncRNAlyzr is available from: https://lncrnalyzr.maayanlab.cloud.
Collapse
Affiliation(s)
- John Erol Evangelista
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Tahleel Ali-Nasser
- Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel.
| | - Lauren E Malek
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Zhuorui Xie
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Giacomo B Marino
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Assaf C Bester
- Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel.
| | - Avi Ma'ayan
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| |
Collapse
|
3
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025; 955:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [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: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
4
|
Chen S, Cheng J, Liu S, Shan D, Wang T, Wang X. Urinary exosomal lnc-TAF12-2:1 promotes bladder cancer progression through the miR-7847-3p/ASB12 regulatory axis. Genes Dis 2025; 12:101384. [PMID: 40297540 PMCID: PMC12036056 DOI: 10.1016/j.gendis.2024.101384] [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: 11/10/2023] [Revised: 06/02/2024] [Accepted: 06/22/2024] [Indexed: 04/30/2025] Open
Abstract
Exosomes encompass a great deal of valuable biological information and play a critical role in tumor development. However, the mechanism of exosomal lncRNAs remains poorly elucidated in bladder cancer (BCa). In this study, we identified exosomal lnc-TAF12-2:1 as a novel biomarker in BCa diagnosis and aimed to investigate the underlying biological function. Dual luciferase reporter assay, RNA immunoprecipitation (RIP), RNA pulldown assays, and xenograft mouse model were used to verify the competitive endogenous RNA mechanism of lnc-TAF12-2:1. We found exosomal lnc-TAF12-2:1 up-regulated in urinary exosomes, tumor tissues of patients, and BCa cells. Down-regulation of lnc-TAF12-2:1 impaired BCa cell proliferation and migration, and promoted cell cycle arrest at the G0/G1 phase and cell apoptosis. The opposite effects were also observed when lnc-TAF12-2:1 was overexpressed. lnc-TAF12-2:1 was transferred by intercellular exosomes to modulate malignant biological behavior. Mechanistically, lnc-TAF12-2:1 packaged in the exosomes relieved the miRNA-mediated silence effect on ASB12 via serving as a sponger of miR-7847-3p to accelerate progression in BCa. ASB12 was also first proved as an oncogene to promote cell proliferation and migration and depress cell cycle arrest and cell apoptosis in our data. In conclusion, exosomal lnc-TAF12-2:1, located in the cytoplasm of BCa, might act as a competitive endogenous RNA to competitively bind to miR-7847-3p, and then be involved in miR-7847-3p/ASB12 regulatory axis to promote tumorigenesis, which provided a deeper insight into the molecular mechanism of BCa.
Collapse
Affiliation(s)
- Song Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Institute of Urology, Wuhan University, Wuhan, Hubei 430071, China
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, Hubei 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, Hubei 430071, China
| | - Jie Cheng
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Shuangtai Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Institute of Urology, Wuhan University, Wuhan, Hubei 430071, China
| | - Danni Shan
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, Hubei 430071, China
| | - Ting Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Institute of Urology, Wuhan University, Wuhan, Hubei 430071, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Institute of Urology, Wuhan University, Wuhan, Hubei 430071, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, Hubei 430071, China
- Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, Hubei 430071, China
- TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
5
|
Alkhathami AG, Altalbawy FMA, Rizaev JA, H M, Jaswinder Kaur, Ali Jeddoa ZM, Jabir MS, Jawad SF, Yumashev A, Zwamel AH. An overview of lncRNA GAPLINC's role in human cancer growth and metastasis. Arch Biochem Biophys 2025; 771:110506. [PMID: 40513977 DOI: 10.1016/j.abb.2025.110506] [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: 04/25/2025] [Revised: 06/05/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as critical regulators in cancer biology, influencing tumor initiation, progression, and metastasis. Among them, GAPLINC (Gastric Adenocarcinoma Predictive Long Intergenic Non-coding RNA) has garnered attention due to its aberrant expression across various malignancies and its association with poor clinical outcomes. While several studies have highlighted GAPLINC's oncogenic roles through interactions with microRNAs and involvement in epithelial-mesenchymal transition (EMT), a comprehensive synthesis of its mechanistic functions and clinical implications remains limited. This review addresses this limitation by systematically analyzing current findings on GAPLINC's molecular mechanisms, regulatory networks, and functional roles in different cancer types, including gastric, colorectal, non-small cell lung cancer (NSCLC), glioma, renal cell carcinoma (RCC), and esophageal squamous cell carcinoma (ESCC). We further evaluate the potential of GAPLINC as a diagnostic, prognostic, and therapeutic target, providing an integrated perspective that consolidates existing knowledge and identifies directions for future research. By highlighting both common and cancer-specific mechanisms involving GAPLINC, this review adds new insights into its significance as a versatile biomarker and therapeutic target in oncology.
Collapse
Affiliation(s)
- Ali G Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Jasur Alimdjanovich Rizaev
- Department of Public health and Healthcare management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Malathi H
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Jaswinder Kaur
- Chandigarh pharmacy college, Chandigarh Group of colleges, jhanjeri, Mohali 140307, punjab, India
| | | | - Majid S Jabir
- Department of applied sciences/ university of technology- Iraq
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, 51001 Hillah, Babylon, Iraq
| | | | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq*; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
6
|
Chen Y, Zhao N, Xu L, Jia X, Liu F, Huang J, Li X, Wang Y, Lai C, Shen Y, Wang F, Lv Y, Huang X, Zhang F, Gu H, Dai S. Integrative multi-omics analysis reveals the LncRNA 60967.1-PLCD4-ATRA axis as a key regulator of colorectal cancer progression and immune response. Mol Cancer 2025; 24:164. [PMID: 40481569 PMCID: PMC12142938 DOI: 10.1186/s12943-025-02359-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 05/19/2025] [Indexed: 06/11/2025] Open
Abstract
Colorectal cancer (CRC) is a major global health concern, characterized by high morbidity and mortality rates. CRC progression involves intricate molecular networks that remain incompletely understood. In this study, we conducted an integrative multi-omics analysis of transcriptomic, proteomic, and metabolomic profiles from CRC tissues and matched normal adjacent tissues (NATs). Our analysis revealed 1,394 differentially expressed long non-Coding RNAs (lncRNAs), 2,788 genes, 548 proteins, and 91 metabolites. A significant interaction network comprising 22 lncRNAs, 14 mRNAs/proteins, and 9 metabolites was identified, among which lncRNA 60967.1 emerged as a pivotal regulator. Functional validation demonstrated that lncRNA 60967.1 is markedly downregulated in CRC cell lines and patient tissues. Overexpression of lncRNA 60967.1 restored expression of the tumor suppressor PLCD4 and increased levels of all-trans retinoic acid (ATRA). This modulation enhanced IFN-γ-induced apoptosis and increased expression of the IFN-γ receptor subunit IFNGR1, thereby partially reversing IFN-γ resistance. In murine models, lncRNA 60967.1 overexpression promoted immune cell infiltration and synergized with anti-PD-1 therapy to inhibit tumor growth. Collectively, our findings uncover a novel lncRNA-mRNA/protein-metabolite network, the lncRNA 60967.1-PLCD4-ATRA axis, that plays a critical role in CRC progression and immune modulation, offering promising therapeutic targets for improved treatment efficacy.
Collapse
Affiliation(s)
- Yiyi Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Ningning Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China.
| | - Lingna Xu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Xiya Jia
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Fang Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China
| | - Jian Huang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China
| | - Xuhua Li
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China
| | - Yunfei Wang
- Hangzhou ShengTing Medical Technology Co., Ltd, Hangzhou, Zhejiang Province, 310018, China
| | - Chuanxi Lai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Yanbin Shen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Fei Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Yiming Lv
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Fan Zhang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China.
| | - Hongcang Gu
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China.
| | - Sheng Dai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
7
|
Xu X, Mao X, Liu W, Ming Y, Zhang T, Yang Y, Gu‐Ha A, Lin Y, Peng Y. The Long Noncoding RNA LINC02820 Promotes Tumor Growth and Metastasis Through Regulating MYH9 Expression in Esophageal Squamous Cell Carcinoma. MedComm (Beijing) 2025; 6:e70218. [PMID: 40416600 PMCID: PMC12099067 DOI: 10.1002/mco2.70218] [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: 04/14/2024] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) play important roles in tumorigenesis, but their biological functions and mechanisms in esophageal squamous cell carcinoma (ESCC) remain poorly understood. In this study, we employed high-throughput sequencing and bioinformatics analyses to identify the differentially expressed lncRNAs between ESCC tumors and adjacent normal tissues, among which LINC02820 is significantly upregulated in ESCC. Rapid amplification of cDNA ends assays determined the transcription initiation and termination sites of LINC02820, confirming it as a novel transcript variant localized in both the nucleus and cytoplasm of ESCC cells. Functional studies demonstrated that LINC02820 promotes cell proliferation and migration in vitro and enhances tumor growth and metastasis in vivo. Mechanistically, LINC02820 interacts with Myosin-9 protein and prevent it from ubiquitination-mediated proteasomal degradation. Additionally, the RNA-binding protein insulin-like growth factor 2 mRNA-binding protein 2 binds to LINC02820 and increase its RNA stability in ESCC cells, thus upregulating LINC02820 expression. Therefore, these findings indicate LINC02820 as an oncogenic lncRNA in ESCC progression and suggest its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xiaomin Xu
- Center for Molecular OncologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Xinting Mao
- Center for Molecular OncologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Wenrong Liu
- Center for Molecular OncologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yue Ming
- Center for Molecular OncologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Tingting Zhang
- Center for Molecular OncologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yang Yang
- Center for Molecular OncologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - A‐Lai Gu‐Ha
- Department of Thoracic SurgeryWest China HospitalSichuan UniversityChengduChina
| | - Yi‐Dan Lin
- Department of Thoracic SurgeryWest China HospitalSichuan UniversityChengduChina
| | - Yong Peng
- Center for Molecular OncologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
8
|
Salim Abed H, Oghenemaro EF, Kubaev A, Jeddoa ZMA, S R, Sharma S, Vashishth R, Jabir MS, Jawad SF, Zwamel AH. Non-coding RNAs as a Critical Player in the Regulation of Inflammasome in Inflammatory Bowel Diseases; Emphasize on lncRNAs. Cell Biochem Biophys 2025; 83:1359-1374. [PMID: 39424765 DOI: 10.1007/s12013-024-01585-2] [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] [Accepted: 09/28/2024] [Indexed: 10/21/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic disease caused by a dysregulated immune response to host intestinal microflora. A hyperactive inflammatory and immunological response in the gut has been shown to be one of the disease's long-term causes despite the complexity of the clinical pathology of IBD. The innate immune system activator known as human gut inflammasome is thought to be a significant underlying cause of pathology and is closely linked to the development of IBD. It is essential to comprehend the function of inflammasome activation in IBD to treat it effectively. Systemic inflammasome regulation may be a proper therapeutic and clinical strategy to manage IBD symptoms since inflammasomes may have a significant function in IBD. Non-coding RNAs (ncRNAs) are a type of RNA transcript that is incapable of encoding proteins or peptides. In IBD, inflammation develops and worsens as a result of its imbalance. Culminating evidence has been shown that ncRNAs, and particularly long non-coding RNAs (lncRNAs), may play a role in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in IBD. The relationship between IBD and the gut inflammasome, as well as current developments in IBD research and treatment approaches, have been the main topics of this review. We have covered inflammasomes and their constituents, results from in vivo research, inflammasome inhibitors, and advancements in inflammasome-targeted therapeutics for IBD.
Collapse
Affiliation(s)
- Hussein Salim Abed
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Ramadi, Iraq
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria.
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, 140100, Uzbekistan
| | | | - RenukaJyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Shilpa Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Karbala, Iraq
| | - Sabrean Farhan Jawad
- Department of Biochemistry, College of Science, Al-Mustaqbal University, 51001, Babil, Iraq
| | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
9
|
Castro-Oropeza R, Velazquez-Velazquez C, Vazquez-Santillan K, Mantilla-Morales A, Ruiz Tachiquin ME, Torres J, Rios-Sarabia N, Mayani H, Piña-Sanchez P. Landscape of lncRNAs expressed in Mexican patients with triple‑negative breast cancer. Mol Med Rep 2025; 31:163. [PMID: 40211710 PMCID: PMC12015155 DOI: 10.3892/mmr.2025.13528] [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: 07/31/2024] [Accepted: 02/24/2025] [Indexed: 04/25/2025] Open
Abstract
Long non‑coding RNAs (lncRNAs) are key regulators of gene expression, that can regulate a range of carcinogenic processes. Moreover, they exhibit stability in biological fluids, with some displaying tissue specificity. As their expression depends on specific conditions or is linked to the regulation of particular signaling pathways, lncRNAs are promising candidates for providing insights into the likely progression of the disease. This allows for the stratification of patients based on their risk of progression, making them potential prognostic biomarkers in various types of cancer. In addition, the tissue‑specific expression profile of lncRNAs renders them ideal candidates for detection, prognosis and monitoring of cancer progression. The present study aims to provide an overview of differentially expressed lncRNAs in Mexican patients with triple‑negative breast cancer (TNBC), a subtype of breast cancer. The aim was to identify potential prognostic biomarkers that can be applied to improve the clinical management of Mexican patients with TNBC. Human Transcriptome Array 2.0 microarrays were used to analyze the transcriptome of TNBC and luminal tumors, which are reported to have a good prognosis amongst aggressive tumor types. Subsequently, results from these microarrays were validated in a cohort from The Cancer Genome Atlas, an independent cohort of Mexican patients and in breast cancer cell lines (MCF7, ZR75, T47D, MDA‑MB‑231, MDA‑MB‑468 and BT20). A total of 746 differentially expressed transcripts were identified, including 102 lncRNAs in TNBC compared with luminal tumors. Among the lncRNAs with the most significant changes in expression levels, SOX9‑AS was highly expressed in TNBC, whereas the expression of Lnc‑peroxidasin‑3:1 (Lnc‑PXDN‑3:1), Lnc‑RNA Synapse Defective Rho GTPase Homolog (Lnc‑SYDE) and long intergenic non‑coding RNA (LINC)01087 were decreased. In addition, the low expression of lncRNA LINC01087, LINC02568, ACO22196, and lncRNA eosinophil granule ontogeny transcript (Lnc‑EGOT) was associated with poor overall survival (OS). Further analysis revealed that the high expression levels of Lnc‑PXDN‑3:1, Lnc RNA fibrous sheath interacting protein 1‑6:3 and (LINC)00182 were associated with reduced survival in patients with the luminal subtype of breast cancer. Similarly, low expression levels of lncRNAs such as GATA binding protein 3‑1 (Lnc‑GATA‑3‑1), LINC01087, and BX679671.1 in luminal subtypes of breast cancer, as well as LINC00504 and LncRNA rho guanine nucleotide exchange factor 38 intronic transcript 1 (Lnc‑ARHGEF38‑IT1) in basal subtypes have been linked to poorer survival. The interactions and functions of LINC01087 were then investigated, revealing the interaction of LINC01087 with RNAs and transcription factors, highlighting their potential involvement in the estrogen receptor pathway. The present study provided a detailed analysis of the expression of lncRNAs in TNBC, which highlights the role of lncRNAs as a biomarker in the survival outcomes of patients with breast cancer to improve the understanding of transcriptional regulation in TNBC.
Collapse
Affiliation(s)
- Rosario Castro-Oropeza
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Cindy Velazquez-Velazquez
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Karla Vazquez-Santillan
- Laboratory of Innovation in Precision Medicine, National Institute of Genomic Medicine, Mexico City 14610, Mexico
| | - Alejandra Mantilla-Morales
- Department of Pathology, High Specialty Medical Unit Oncology Hospital, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Martha-Eugenia Ruiz Tachiquin
- Molecular Biology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Javier Torres
- Infectious and Parasitic Diseases Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Nora Rios-Sarabia
- Infectious and Parasitic Diseases Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Hector Mayani
- Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Patricia Piña-Sanchez
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| |
Collapse
|
10
|
Qi H, Ying G, Ling W, Jia H, Zhou X, Lin X. The role of lncRNAs in sepsis-induced acute lung injury: Molecular mechanisms and therapeutic potential. Arch Biochem Biophys 2025; 768:110407. [PMID: 40180295 DOI: 10.1016/j.abb.2025.110407] [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: 03/02/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Sepsis, a life-threatening syndrome, results from a dysregulated immune and hemostatic response, contributing to acute lung injury (ALI) and its progression into acute respiratory distress syndrome (ARDS). The development of septic ALI is complex, involving excessive inflammatory mediator production that damages endothelial and epithelial cells, leading to vascular leakage, edema, and vasodilation-key factors in ALI pathogenesis. Long noncoding RNAs (lncRNAs), over 200 nucleotides in length, play critical roles in various biological processes, including sepsis regulation. They exhibit both promotive and inhibitory effects, influencing sepsis progression and resolution. Despite their significance, comprehensive reviews detailing lncRNA involvement in sepsis-induced ALI remain limited. This review aims to address this gap by summarizing the diverse functions of lncRNAs in septic ALI, emphasizing their potential in diagnosis and treatment. Furthermore, we will explore the molecular mechanisms underlying lncRNA involvement, particularly their miRNA-dependent regulatory pathways. Understanding these interactions may provide novel insights into therapeutic strategies for sepsis-induced ALI.
Collapse
Affiliation(s)
- Huijuan Qi
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China.
| | - Gu Ying
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| | - Wang Ling
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| | - Honggang Jia
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| | - Xinxiu Zhou
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| | - Xinyu Lin
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| |
Collapse
|
11
|
Che R, Panah M, Mirani B, Knowles K, Ostapovich A, Majumdar D, Chen X, DeSimone J, White W, Noonan M, Luo H, Alexandrov A. Identification of human pathways acting on nuclear non-coding RNAs using the Mirror forward genetic approach. Nat Commun 2025; 16:4741. [PMID: 40399278 PMCID: PMC12095793 DOI: 10.1038/s41467-025-59998-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/09/2025] [Indexed: 05/23/2025] Open
Abstract
Despite critical roles in diseases, human pathways acting on strictly nuclear non-coding RNAs have been refractory to forward genetics. To enable their forward genetic discovery, we developed a single-cell approach that "Mirrors" activities of nuclear pathways with cytoplasmic fluorescence. Application of Mirror to two nuclear pathways targeting MALAT1's 3' end, the pathway of its maturation and the other, the degradation pathway blocked by the triple-helical Element for Nuclear Expression (ENE), identified nearly all components of three complexes: Ribonuclease P and the RNA Exosome, including nuclear DIS3, EXOSC10, and C1D, as well as the Nuclear Exosome Targeting (NEXT) complex. Additionally, Mirror identified DEAD-box helicase DDX59 associated with the genetic disorder Oral-Facial-Digital syndrome (OFD), yet lacking known substrates or roles in nuclear RNA degradation. Knockout of DDX59 exhibits stabilization of the full-length MALAT1 with a stability-compromised ENE and increases levels of 3'-extended forms of small nuclear RNAs. It also exhibits extensive retention of minor introns, including in OFD-associated genes, suggesting a mechanism for DDX59 association with OFD. Mirror efficiently identifies pathways acting on strictly nuclear non-coding RNAs, including essential and indirectly-acting components, and as a result can uncover unexpected links to human disease.
Collapse
Affiliation(s)
- Rui Che
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
- Clemson University Center for Human Genetics, Greenwood, SC, USA
| | - Monireh Panah
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
- Clemson University Center for Human Genetics, Greenwood, SC, USA
| | - Bhoomi Mirani
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
- Clemson University Center for Human Genetics, Greenwood, SC, USA
| | - Krista Knowles
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
- Clemson University Center for Human Genetics, Greenwood, SC, USA
| | - Anastacia Ostapovich
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Debarati Majumdar
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
- Clemson University Center for Human Genetics, Greenwood, SC, USA
| | - Xiaotong Chen
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
| | - Joseph DeSimone
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
| | - William White
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
| | - Megan Noonan
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
| | - Hong Luo
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
| | - Andrei Alexandrov
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA.
- Clemson University Center for Human Genetics, Greenwood, SC, USA.
| |
Collapse
|
12
|
Obanya DI, Wootton LM, Morgan EL. Advances in understanding the mechanisms of the human papillomavirus oncoproteins. Biochem Soc Trans 2025:BST20253041. [PMID: 40380881 DOI: 10.1042/bst20253041] [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: 02/13/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
High-risk human papillomaviruses (HPVs) are responsible for almost all cervical cancer cases and a growing number of oropharyngeal and anogenital cancers. The primary HPV oncoproteins, E6 and E7, act together to manipulate multiple cellular pathways that can ultimately result in malignant transformation. This includes the deregulation of several signalling pathways that regulate cell proliferation, cell cycle progression and cell survival. Although multiple functions of HPV E6 and E7 in driving oncogenesis are well known, recent studies have uncovered novel oncogenic functions of the HPV oncoproteins, including the manipulation of emerging mechanisms of cancer development, such as epigenetic modifications, cellular plasticity and genomic instability. This review explores current advances in understanding how the HPV oncoproteins interact with these cellular processes, highlighting potential therapeutic targets in HPV-associated cancers.
Collapse
Affiliation(s)
| | | | - Ethan L Morgan
- School of Life Sciences, University of Sussex, Brighton, U.K
| |
Collapse
|
13
|
Liu X, Haugh W, Zhang Z, Huang J. Emerging Role of Long, Non-Coding RNA Nuclear-Enriched Abundant Transcript 1 in Stress- and Immune-Related Diseases. Int J Mol Sci 2025; 26:4413. [PMID: 40362651 PMCID: PMC12072541 DOI: 10.3390/ijms26094413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 05/03/2025] [Accepted: 05/04/2025] [Indexed: 05/15/2025] Open
Abstract
Long, non-coding RNAs (lncRNAs) are a class of RNAs exceeding 200 nucleotides in length, lacking the ability to be translated into proteins. Over the past few decades, an increasing number of publications have established lncRNAs as potent regulators in a broad spectrum of diseases. They modulate the expression of critical genes by affecting transcription, post-transcription, translation, and protein modification. This regulation frequently involves the interaction of lncRNAs with various molecules, such as proteins, RNA, and DNA. lncRNAs are involved in diseases where stress is a significant factor. In recent years, lncRNAs have been identified as regulators of both innate and adaptive immune responses, playing significant roles in the onset and progression of diseases. Additionally, lncRNAs hold potential as biomarkers or therapeutic targets for numerous stress- and immune-related diseases. lncRNA nuclear-enriched abundant transcript 1 (NEAT1) is a notable example. This review consolidates the latest findings about the role of lncRNA NEAT1 in stress response and immune cell function in non-cancer diseases. It summarizes studies on NEAT1 regulating stress response, both innate and adaptive immunity, and its potential as a biomarker and therapeutic target for stress- and immune-related diseases.
Collapse
Affiliation(s)
- Xingliang Liu
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR 97213, USA; (X.L.); (W.H.)
| | - William Haugh
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR 97213, USA; (X.L.); (W.H.)
| | - Ziqiang Zhang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jianguo Huang
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR 97213, USA; (X.L.); (W.H.)
| |
Collapse
|
14
|
Liu Q, Xiong W. LINC00534 promotes breast cancer progression by targeting the miR-139-5p/HMGB2 axis. Discov Oncol 2025; 16:655. [PMID: 40314851 PMCID: PMC12048369 DOI: 10.1007/s12672-025-02483-6] [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: 02/13/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Breast cancer is the most prevalent malignancy among women, it is crucial to identify sensitive biomarkers for prognosis and treatment of breast cancer patients. Emerging research has demonstrated the involvement of long noncoding RNAs (lncRNAs) in the advancement of breast cancer. LINC00534 has recently emerged as a potential regulator in multiple malignancies, yet its clinical significance and molecular mechanisms in breast cancer remain poorly characterized. OBJECTIVE The purpose of this study was to explore the function of LINC00534 and miR-139-5p in breast cancer progression, as well as the mechanisms that underpin its actions. METHODS Tumor and normal tissues were collected from 80 breast cancer patients. qRT-PCR was performed to detect LINC00534 expression in tissues. Kaplan-Meier analysis was used to assess survival differences between groups and the correlation between LINC00534 expression and clinical outcomes. CCK-8 assay was used to evaluate cell proliferation to assess LINC00534's effect on tumor growth. To evaluate the impact of LINC00534 on tumor metastasis, transwell assay was used to detect the migration and invasion abilities of cells. Moreover, dual-luciferase assay was used to verify the relationship within the LINC00534/miR-139-5p/HMGB2 axis. RESULT LINC00534 was significantly upregulated in breast cancer tumor tissues and cell lines (p < 0.001). Higher LINC00534 expression correlated with poorer prognosis in breast cancer patients, including shorter survival and higher recurrence risk (Log-rank p = 0.014). Furthermore, LINC00534 promoted breast cancer cell proliferation, migration, and invasion (all p < 0.001) via its interaction with the miR-139-5p/HMGB2 axis. CONCLUSION LINC00534 may serve as a prognostic marker and the LINC00534/miR-139-5p/HMGB2 axis could be a therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Qiaomei Liu
- Pain Rehabilitation Department, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Xiong
- Department of Thyroid and Breast Surgery, Hubei Provincial Hospital of TCM, Hubei Shizhen Laboratory, No. 4 Garden Hill, Rouge Road, Wuchang District, Wuhan, 430061, China.
| |
Collapse
|
15
|
Chao YL, Zhou KI, Forbes KK, Porrello A, Gentile GM, Zhu Y, Chack AC, John Mary DJS, Liu H, Cockman E, Edatt L, Goda GA, Zhao JJ, Abou Assi H, Wiedner HJ, Tsai Y, Wilkinson L, Van Swearingen AED, Carey LA, Giudice J, Dominguez D, Holley CL, Pecot CV. Snord67 promotes breast cancer metastasis by guiding U6 modification and modulating the splicing landscape. Nat Commun 2025; 16:4118. [PMID: 40316533 PMCID: PMC12048515 DOI: 10.1038/s41467-025-59406-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Previously considered "housekeeping" genes, small nucleolar RNAs (snoRNAs) are increasingly understood to have wide-ranging functions in cancer, yet their role in metastasis has been less well studied. Here, we identify the snoRNA Snord67 as a regulator of lymph node (LN) metastasis in breast cancer. Snord67 expression is enriched in LN metastases in an immune-competent mouse model of female breast cancer. In an orthotopic breast cancer model, loss of Snord67 decreases LN metastasis. In a model of lymphatic metastasis, Snord67 loss decreases LN tumor growth and distant metastases. In breast cancer cell lines, Snord67 knockout results in loss of targeted 2'-O-methylation on U6 small nuclear RNA, as well as widespread changes in splicing. Together, these results demonstrate that Snord67 regulates splicing and promotes the growth of LN metastases and subsequent spread to distant metastases. SnoRNA-guided modifications of the spliceosome and regulation of splicing may represent a potentially targetable pathway in cancer.
Collapse
Affiliation(s)
- Yvonne L Chao
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
- VA Pittsburgh Health System, Pittsburgh, PA, USA
| | - Katherine I Zhou
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kwame K Forbes
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alessandro Porrello
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle M Gentile
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yinzhou Zhu
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Aaron C Chack
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dixcy J S John Mary
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haizhou Liu
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Cockman
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Lincy Edatt
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Grant A Goda
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Justin J Zhao
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hala Abou Assi
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hannah J Wiedner
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yihsuan Tsai
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Lily Wilkinson
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | | | - Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jimena Giudice
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel Dominguez
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA.
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Christopher L Holley
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Chad V Pecot
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA.
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
16
|
Xiao H, Zhou T, Yang Y, Yang X, Bi Y, Cheng X. LncRNA-DANCR Promotes ESCC Progression and Function as ceRNA to Regulate DDIT3 Expression by Sponging microRNA-3193. Cancer Sci 2025; 116:1324-1338. [PMID: 40071783 PMCID: PMC12044675 DOI: 10.1111/cas.70035] [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: 10/22/2024] [Revised: 02/13/2025] [Accepted: 02/22/2025] [Indexed: 05/02/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as crucial regulators of cancer development and progression. Among them, Differentiation Antagonizing Non-Protein Coding RNA (DANCR) has been implicated in various malignancies, including esophageal squamous cell carcinoma (ESCC). This study explores the clinical characteristics, prognostic implications, functional roles, and molecular mechanisms of DANCR in ESCC. Our results demonstrate that DANCR is highly expressed in ESCC, and acts as an oncogene in ESCC both in vitro and in vivo. Through bioinformatics analysis and experimental validation, we revealed that DANCR promotes ESCC progression by sponging miR-3193 and regulating its target gene DDIT3 expression. These findings highlight the critical role of DANCR in the development of ESCC and suggest its potential as a prognostic predictor and drug therapeutic target.
Collapse
Affiliation(s)
- Heng Xiao
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal CancerShanxi Medical UniversityTaiyuanShanxiChina
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of PathologyShanxi Medical UniversityTaiyuanShanxiChina
| | - Tong Zhou
- Shanxi Academy of Medical ScienceShanxi Medical UniversityTaiyuanChina
| | - Yanfang Yang
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal CancerShanxi Medical UniversityTaiyuanShanxiChina
- The School of Public HealthBaotou Medical CollegeBaotouInner MongoliaChina
| | - Xin Yang
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal CancerShanxi Medical UniversityTaiyuanShanxiChina
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of PathologyShanxi Medical UniversityTaiyuanShanxiChina
| | - Yanghui Bi
- Center of Gene Sequencing, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Xiaolong Cheng
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal CancerShanxi Medical UniversityTaiyuanShanxiChina
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of PathologyShanxi Medical UniversityTaiyuanShanxiChina
| |
Collapse
|
17
|
Liu Y, Li X, Wang F, Cai J, Li Z, Huang Y, Duan X, Liu X, He Y, Xu G, Lu Q. MTA1-DT promotes endometrial cancer growth by modulating G2/M-related gene transcription via PURα. Int J Biol Macromol 2025; 309:142943. [PMID: 40210047 DOI: 10.1016/j.ijbiomac.2025.142943] [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: 03/05/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
In recent years, patients with early endometrial cancer (EC) can achieve a good prognosis through surgery. However, advanced and recurrent cases have still posed significant therapeutic challenges. This study aimed to investigate the biological function of long non-coding RNAs (lncRNAs) in EC and elucidate its underlying molecular mechanism. Through quantitative reverse transcription polymerase chain reaction (RT-qPCR) analysis, functional assays in cell lines, and bioinformatics approaches, we identified lncRNA MTA1-DT as a novel oncogenic factor in EC progression. RNA-seq and RT-qPCR analysis demonstrated that MTA1-DT was significantly upregulated with a 5-fold increase in EC cell lines compared to normal controls. Functional studies revealed that MTA1-DT promoted cell proliferation and migration. Mechanistically, we demonstrated that MTA1-DT physically interacted with purine-rich element binding protein-alpha (PURα) and facilitated its nuclear translocation, thereby enhancing its transcription factor activity. This nuclear accumulation of PURα promoted the transcription of downstream G2/M related genes, particularly EGF, leading to accelerated tumor growth. Thus, these results indicate that MTA1-DT exerts its oncogenic effects in EC through regulation of the cell cycle. Our findings establish MTA1-DT as a promising therapeutic target for EC treatment and provide new insights into the molecular mechanisms underlying EC progression.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Xin Li
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
| | - Fanchen Wang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jinhui Cai
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Zhouqi Li
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Yanchun Huang
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Xiaoling Duan
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Xinyi Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Yuxin He
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China.
| | - Qi Lu
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China.
| |
Collapse
|
18
|
Chen M, Feng X, Liu C, Huang Y, Su L, Li X, Zhu J. Diagnostic value of exosome-derived lncRNA PITPNA-AS1 in lung cancer. Front Immunol 2025; 16:1539557. [PMID: 40342419 PMCID: PMC12058797 DOI: 10.3389/fimmu.2025.1539557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/25/2025] [Indexed: 05/11/2025] Open
Abstract
Background Lung cancer is one of the most lethal types of cancer, and effective diagnostic biomarkers are required. There is increasing evidences that exosome-secreted lncRNAs could play an important role in lung cancer diagnosis. However, the diagnostic value and molecular mechanism of the key lncRNA PITPNA-AS1 in lung cancer remain unclear. Methods qRT-PCR was conducted to determine the levels of exosomal lncRNA PITPNA-AS1 in pleural effusions from lung adenocarcinoma, squamous cell lung carcinoma, and small cell lung cancer patients. Receiver operating characteristic (ROC) curve analyses were used to evaluate the diagnostic accuracy of PITPNA-AS1. Its role in lung cancer development was determined by a series of experiments, including CCK-8, flow cytometry, and transwell assays. RNA pull-down and RNA immunoprecipitation assays were carried out to examine the interaction between PITPNA-AS1 and Fragile X messenger ribonucleoprotein 1 (FMR1). Results We discovered PITPNA-AS1 in exosomes from lung cancer patients. Its expression was significantly increased in lung cancer patients compared to non-cancer patients, and it was strongly associated with tumor stage, lymph node metastasis, and distant metastasis in all lung cancer subtypes assessed (all p<0.05). ROC curve analyses demonstrated that exosomal PITPNA-AS1 had a high accuracy for differentiating among lung cancer subtypes. Furthermore, PITPNA-AS1 boosted H1299 and A549 cell proliferation, migration, and invasion. Mechanistically, via direct interaction, PITPNA-AS1 increased FMR1 stability by preventing its ubiquitination. Conclusions These results reveal that exosome-derived lncRNA PITPNA-AS1 acts as an oncogene to promote malignant biological behaviors and is a promising diagnostic biomarker in lung cancer.
Collapse
Affiliation(s)
- Mujin Chen
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| | - XiaoHui Feng
- Department of Oncology, Loujiang New City Hospital of Taicang (Ruijin Hospital, Shanghai Jiao Tong University School of Medicine), Suzhou, China
| | - ChengChen Liu
- Department of Gastroenterology, WuWei City The Second People’s Hospital, Gansu, China
| | - Yan Huang
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| | - LiJuan Su
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| | - XiaoFeng Li
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| | - JinFeng Zhu
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| |
Collapse
|
19
|
Ibarra-Sierra E, Bermúdez M, Villegas-Mercado CE, Silva-Cázares MB, López-Camarillo C. LncRNAs Regulate Vasculogenic Mimicry in Human Cancers. Cells 2025; 14:616. [PMID: 40277941 PMCID: PMC12025595 DOI: 10.3390/cells14080616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025] Open
Abstract
Vasculogenic mimicry (VM) has recently been discovered as an alternative mechanism for nourishing cancer cells in vivo. During VM, tumor cells align and organize themselves into three-dimensional (3D) channel-like structures to transport nutrients and oxygen to the internal layers of tumors. This mechanism mainly occurs in aggressive solid tumors and has been associated with poor prognosis in oncologic patients. Long non-coding RNAs (lncRNAs) are essential regulators of protein-encoding genes involved in cancer development and progression. These single-stranded RNA molecules regulate critical cellular functions in cancer cells including cell proliferation, apoptosis, angiogenesis, VM, therapy response, migration, invasion, and metastasis. Recently, high-throughput RNA-sequencing technologies have identified thousands of lncRNAs, but only a small percentage of them have been functionally characterized in human cancers. The vast amount of data about its genomic expression in tumors can allow us to dissect their functions in cancer biology and make them suitable biomarkers for cancer diagnosis and prognosis. In this study, we reviewed the current knowledge about the role of lncRNAs in regulating VM in cancer. We also examined the molecular mechanisms of lncRNAs and highlight several commonalities in the cellular functions associated with VM between diverse cancer types. Future directions for research focused on deciphering their function in VM are delineated. Finally, the potential of selected lncRNAs as novel therapeutic targets in RNA-based molecular interventions is also discussed.
Collapse
Affiliation(s)
- Eloísa Ibarra-Sierra
- Instituto Estatal de Cancerología “Dr. Arturo Beltrán Ortega”, Acapulco Guerrero 39530, Mexico;
| | - Mercedes Bermúdez
- Facultad de Odontología, Universidad Autónoma de Chihuahua, Chihuahua 31000, Mexico; (M.B.); (C.E.V.-M.)
| | | | - Macrina B. Silva-Cázares
- Unidad Académica Multidisciplinaria Región Altiplano, Universidad Autónoma de San Luis Potosí, Matehuala 78700, Mexico;
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México 03100, Mexico
| |
Collapse
|
20
|
Mouhou E, Genty F, El M'selmi W, Chouali H, Zagury JF, Le Clerc S, Proudhon C, Noirel J. High tissue specificity of lncRNAs maximises the prediction of tissue of origin of circulating DNA. Sci Rep 2025; 15:12941. [PMID: 40234550 PMCID: PMC12000428 DOI: 10.1038/s41598-024-82393-9] [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: 03/21/2024] [Accepted: 12/05/2024] [Indexed: 04/17/2025] Open
Abstract
Several studies have made it possible to envision a translational application of plasma DNA sequencing in cancer diagnosis and monitoring. However, the extremely low concentration of circulating tumour DNA (ctDNA) fragments among the total cell-free DNA (cfDNA) remains a formidable challenge to overcome and statistical models have yet to be improved enough to become of practical use. In this study, we set about appraising the predictive value of a variety of binary classification models based on cfDNA sequencing using fragmentation features extracted around transcription start sites (TSSs). We investigated (1) features summarising mapped fragment density around each TSS, (2) long non-coding RNA (lncRNA) genes versus coding genes and (3) selection criteria to generate gene classes to be assigned by the model. Given that, in healthy samples, most of the cfDNA comes from lymphomyeloid lineages, we could identify the model parametrisation with the best accuracy in those lineages using publicly available datasets of healthy patients' cfDNA. Our results show that (1) the way tissue-specific gene classes are defined matters more than what fragmentation features are included, and (2) in particular, lncRNAs are more tissue specific than coding genes and stand out in terms of both sensitivity and specificity in our results.
Collapse
Affiliation(s)
- Elyas Mouhou
- Laboratoire GBCM (EA7528), Conservatoire national des arts et métiers (CNAM), Paris, France
| | - Fabien Genty
- Infotel Conseil, 13, rue Madeleine-Michelis, Neuilly-sur-Seine, France
| | | | - Hanae Chouali
- BioinfOmics, GenoToul Bioinformatics facility, Université Fédérale de Toulouse, INRAE, Castanet-Tolosan, France
- MIAT, Université Fédérale de Toulouse, INRAE, Castanet-Tolosan, France
| | - Jean-François Zagury
- Laboratoire GBCM (EA7528), Conservatoire national des arts et métiers (CNAM), Paris, France
| | - Sigrid Le Clerc
- Laboratoire GBCM (EA7528), Conservatoire national des arts et métiers (CNAM), Paris, France
| | | | - Josselin Noirel
- Laboratoire GBCM (EA7528), Conservatoire national des arts et métiers (CNAM), Paris, France.
| |
Collapse
|
21
|
Yang Y, Wang TY, Li Q, Lu J, Ren Y, Weiner AB, Fry J, Liu Q, Yum C, Wang R, Guo Q, Wan Y, Ji Z, Dong X, Lotan TL, Schaeffer EM, Yang R, Cao Q. Androgen receptor-regulated lncRNA PRCAT71 promotes AR signaling through the interaction with KHSRP in prostate cancer. SCIENCE ADVANCES 2025; 11:eadk6989. [PMID: 40203114 PMCID: PMC11980854 DOI: 10.1126/sciadv.adk6989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Mounting evidence indicates that long noncoding RNAs (lncRNAs) play vital roles in tumorigenesis and progression of cancers. However, the functions and regulatory mechanisms of lncRNAs in prostate cancer (PCa) are still largely unknown. In this study, we found an lncRNA, PCa-associated transcript 71 (PRCAT71), highly expressed in metastatic and primary PCa compared to benign prostate tissues. Silencing PRCAT71 inhibited cancerous properties of PCa cells and androgen receptor (AR) signaling. Mechanistically, PRCAT71 acts as a scaffold to recruit K homology (KH)-type splicing regulatory protein (KHSRP) to AR messenger RNA (mRNA) and stabilize AR mRNA, leading to activated AR signaling. KHSRP plays a critical role in PCa progression. PRCAT71 is transcriptionally regulated by AR-driven enhancers, forming a positive regulatory loop between AR and PRCAT71 in PCa. Our study demonstrates a coordinated regulation of AR mRNA by lncRNA PRCAT71 and RNA binding protein KHSRP and provides insight that the PRCAT71-KHSRP-AR axis is a promising therapeutic target for treating PCa.
Collapse
Affiliation(s)
- Yongyong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ting-You Wang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qianru Li
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jiawen Lu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yanan Ren
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Adam B. Weiner
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua Fry
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qi Liu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chaehyun Yum
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rui Wang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qingxiang Guo
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yu Wan
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL 60628, USA
| | - Zhe Ji
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL 60628, USA
| | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Edward M. Schaeffer
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rendong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qi Cao
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
22
|
Zhang S, Lan X, Lei L. LINC01559: roles, mechanisms, and clinical implications in human cancers. Hum Cell 2025; 38:83. [PMID: 40205068 DOI: 10.1007/s13577-025-01218-7] [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: 12/15/2024] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Long intergenic non-protein coding RNA 1559 (LINC01559), a long non-coding RNA (lncRNA) located on chromosome 12p13.1, plays a critical role in the progression of various cancers. The aberrant expression of LINC01559 significantly impacts multiple biological processes in tumor cells, including cell proliferation, epithelial-mesenchymal transition (EMT), migration, invasion, angiogenesis, and cellular stemness. Notably, the expression levels of LINC01559 correlate with the pathological features and prognosis of several cancers, such as pancreatic, breast, and gastric cancers, and it may serve as a diagnostic marker for non-small cell lung cancer. Moreover, the expression of LINC01559 is regulated by various mechanisms and can influence cancer initiation and progression through a competing endogenous RNA (ceRNA) network, where it interacts with a cohort of eight different microRNAs (miRNAs). Additionally, LINC01559 may directly interact with downstream proteins, thereby promoting their functions or enhancing their stability. LINC01559 is also implicated in key signaling pathways associated with cancer development, including the PI3 K/AKT, RAS, and autophagy signaling pathways. Furthermore, it has been linked to drug resistance in breast cancer and hepatocellular carcinoma. This review provides a comprehensive assessment of the clinical implications of dysregulated LINC01559 expression across various cancer types, highlighting its crucial functions and underlying molecular mechanisms in tumorigenesis. Additionally, we present in-depth discussions and propose hypotheses regarding the functional roles of LINC01559 in cancer pathogenesis, while outlining potential research avenues for future exploration of this molecular target.
Collapse
Affiliation(s)
- Shuwen Zhang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Xin Lan
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Ling Lei
- Prevention and Treatment Center, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang, 332000, Jiangxi, China.
| |
Collapse
|
23
|
Wang D, Zhao H, Zhao Y, An X, Shi C, Pan Z, Zheng Q, Wang X, Lu J, Li D. Silencing LINC01547 induces hepatocellular carcinoma cell apoptosis and metastasis inhibition via the ADAR1/FAK and miR-146b-5p/RAC1 axes. Apoptosis 2025; 30:936-954. [PMID: 39904859 DOI: 10.1007/s10495-024-02070-0] [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] [Accepted: 12/26/2024] [Indexed: 02/06/2025]
Abstract
Growing research indicates that long noncoding RNAs (lncRNAs) are pivotal in the development and advancement of hepatocellular carcinoma (HCC). Our research pinpointed LINC01547 as a notable lncRNA that was significantly downregulated in Hep3B cells treated with bufotalin, whereas it exhibited elevated expression levels in HCC tumor tissues. Further study found that silencing LINC01547 markedly suppressed proliferation, induced apoptosis, and inhibited migration and invasion in Hep3B and HepG2 cells. LINC01547 knockdown reduced ADAR1 expression, which led to apoptosis and suppressed metastasis via inhibition of the FAK signaling pathway. Additionally, silencing LINC01547 upregulated miR-146b-5p, which in turn decreased RAC1 levels, further promoting apoptosis and inhibiting metastasis in HCC cells. In vivo, a Hep3B tumor-bearing mouse model confirmed the antitumor effects of LINC01547 silencing. Our findings demonstrate that LINC01547 regulates HCC cell apoptosis and metastasis through the ADAR1/FAK and miR-146b-5p/RAC1 pathways, suggesting that LINC01547 may serve as a biomarker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Dan Wang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, PR China
| | - Huijie Zhao
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Ying Zhao
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Xuejing An
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Chuanqin Shi
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, 255020, PR China
| | - Zhaohai Pan
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Qiusheng Zheng
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Xin Wang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, PR China.
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, PR China.
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, 264003, PR China.
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
| |
Collapse
|
24
|
Feng P, Yang F, Zang D, Bai D, Xu L, Fu Y, You R, Liu T, Yang X. Deciphering the roles of cellular and extracellular non-coding RNAs in chemotherapy-induced cardiotoxicity. Mol Cell Biochem 2025; 480:2177-2199. [PMID: 39485641 PMCID: PMC11961477 DOI: 10.1007/s11010-024-05143-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
Chemotherapy-induced cardiotoxicity is a major adverse effect, driven by multiple factors in its pathogenesis. Notably, RNAs have emerged as significant contributors in both cancer and heart failure (HF). RNAs carry genetic and metabolic information that mirrors the current state of cells, making them valuable as potential biomarkers and therapeutic tools for diagnosing, predicting, and treating a range of diseases, including cardiotoxicity. Over 97% of the genome is transcribed into non-coding RNAs (ncRNAs), including ribosomal RNA (rRNAs), transfer RNAs (tRNAs), and newly identified microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs). NcRNAs function not only within their originating cells but also in recipient cells by being transported through extracellular compartments, referred to as extracellular RNAs (exRNAs). Since ncRNAs were identified as key regulators of gene expression, numerous studies have highlighted their significance in both cancer and cardiovascular diseases. Nevertheless, the role of ncRNAs in cardiotoxicity remains not fully elucidated. The study aims to review the existing knowledge on ncRNAs in Cardio-Oncology and explore the potential of ncRNA-based biomarkers and therapies. These investigations could advance the clinical application of ncRNA research, improving early detection and mitigating of chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Pan Feng
- Baoji Hospital of Traditional Chinese Medicine, Baoji, 721000, China
| | - Fan Yang
- Guang'an Men Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Dongmei Zang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Dapeng Bai
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Liyan Xu
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Yueyun Fu
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Ranran You
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Tao Liu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710068, China.
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China.
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
25
|
Meng LW, Luo ZY, Zhang FQ, Dong YX, Ye C, Zhang W, Wang JJ. Increased expression of an isoform of the long non-coding RNA, lnc37707, is associated with malathion resistance in Bactrocera dorsalis. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 209:106343. [PMID: 40082034 DOI: 10.1016/j.pestbp.2025.106343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/16/2025]
Abstract
The oriental fruit fly, Bactrocera dorsalis (Hendel) is an invasive pest threatening global fruit industries. Field populations of B. dorsalis exhibit complex insecticide resistance, hindering pest control efforts and exacerbating damage. Long non-coding RNAs (lncRNAs) are critical regulators of multiple bioprocess in insects, including insecticide resistance, and have potentials as novel target for pest management. Here, the candidate lncRNAs associated with malathion resistance in B. dorsalis were identified through RNA-seq. One of the isoforms of lnc37707, designated as lnc37707.10, was significantly enriched in the detoxification tissues of malathion-resistant (MR) strain. A specific fragment of lnc37707.10 (sflnc37707) was strongly associated with malathion resistance, and silencing sflnc37707 increased the susceptibility, whereas overexpressing it decreased susceptibility to malathion. Silencing sflnc37707 resulted in the down-regulation of 248 genes, but none of them included the four adjacent genes as its potential target. Instead, pathway analysis revealed significant enrichment of down-regulated genes involved in drug and xenobiotics metabolism, including P450s and GSTs. Bioinformatic analysis suggested a potential regulatory role of miRNA in the function of lnc37707. Further combining silence or overexpression sflnc37707 with miRNA mimic treatment identified that BdGSTd10 (an important gene involved in malathion resistance) and miR-1000 was strongly linked to lnc37707.10. Finally, a ceRNA (competing endogenous RNA) regulatory axis was proposed, where lnc37707.10 might indirectly modulate BdGSTd10 by sponging miR-1000 to regulate the malathion resistance in B. dorsalis. These findings provide a new insight into insecticide resistance and a potential lncRNA target for the sustainable pest management.
Collapse
Affiliation(s)
- Li-Wei Meng
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China.; Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China.
| | - Zong-Yu Luo
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China.; Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China
| | - Fu-Qiang Zhang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China.; Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China
| | - Yi-Xuan Dong
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China.; Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China
| | - Chao Ye
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China.; Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China.
| | - Wei Zhang
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China; Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing 400712, China.
| | - Jin-Jun Wang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China.; Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
26
|
Li X, Yang A, Liu X, Zhang R, Zhou H, Xu S. Genetic association of long non-coding RNA ANRIL polymorphism with the risk of type 2 diabetes mellitus in the Chinese Han population. Diabetol Metab Syndr 2025; 17:108. [PMID: 40148977 PMCID: PMC11951501 DOI: 10.1186/s13098-025-01670-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is closely associated with both environmental and genetic factors, involving multi-gene inheritance. This study examined the association between the polymorphic locus rs10757278 in long non-coding RNA ANRIL and T2DM. METHODS Polymerase chain reaction (PCR) was used to detect the rs10757278 polymorphism in the ANRIL gene. RT-qPCR measured ANRIL expression levels, and logistic regression identified independent risk factors for T2DM. Furthermore, the receiver operating characteristic (ROC) curve was constructed to evaluate the clinical diagnostic value of serum ANRIL levels in diagnosing T2DM. RESULTS The rs10757278 polymorphism of the ANRIL was associated with the development of T2DM. Specifically, the G allele increases the risk of T2DM, and individuals carrying the GG genotype have a higher risk of developing the disease. Significant differences were found in low-density lipoprotein cholesterol (LDL-C), fasting plasma glucose (FPG), and glycated hemoglobin (HbA1c) among T2DM patients with different genotypes of ANRIL rs10757278. The relative FPG and HbA1c levels were relatively lower in individuals with the AA genotype and higher in those with the GG genotype. Moreover, serum ANRIL levels in the T2DM group were lower than in the control group. Body mass index (BMI), the rs10757278 locus, and serum ANRIL levels were independent risk factors for the development of T2DM. The ROC curve showed that serum ANRIL levels have significant clinical diagnostic value for the diagnosis of T2DM. CONCLUSION The rs10757278 polymorphism in ANRIL was strongly associated with the genetic predisposition to T2DM.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Emergency, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, China
| | - Aige Yang
- Department of Endocrinology and Metabolism, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, China
| | - Xiao Liu
- Department of Endocrinology and Metabolism, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, China
| | - Rui Zhang
- Department of Endocrinology and Central Laboratory, The First Hospital of Hebei Medical University, No.89, Donggang Road, 050031, Shijiazhuang, China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang, 050031, China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, 050031, China
| | - Huimin Zhou
- Hebei International Joint Research Center for Brain Science, Shijiazhuang, 050031, China.
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, 050031, China.
- Department of Endocrinology and Metabolism, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Shijiazhuang, 050051, China.
| | - Shunjiang Xu
- Department of Endocrinology and Central Laboratory, The First Hospital of Hebei Medical University, No.89, Donggang Road, 050031, Shijiazhuang, China.
- Hebei International Joint Research Center for Brain Science, Shijiazhuang, 050031, China.
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, 050031, China.
| |
Collapse
|
27
|
Wang S, Guo J, Xian X, Li M, Zhang A, Liu Y, Zhang Y, Chen S, Gu G, Zhang X, Yan D, An M, Pan L, Fu B. Distinct 5-methylcytosine profiles of LncRNA in breast cancer brain metastasis. BMC Cancer 2025; 25:557. [PMID: 40148799 PMCID: PMC11951547 DOI: 10.1186/s12885-025-13948-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Recent studies have identified a complex relationship between methylation patterns and the development of various cancers. Breast cancer (BC) is the second leading cause of cancer mortality among women. Approximately 5-20% of BC patients are at risk of BC brain metastases (BCBM). Although 5-methylcytosine (m5C) has been identified as an important regulatory modifier, its distribution in BCBM is not well understood. This study aimed to investigate the distribution of m5C in BCBM. MATERIALS AND METHODS Samples from BCBM (231-BR cells) and BC (MDA-MB-231 cells) groups were subjected to a comprehensive analysis of the m5C methylation in long non-coding RNA (lncRNA) using methylated RNA immunoprecipitation next-generation sequencing (MeRIP-seq). The expression levels of methylated genes in BC and adjacent tissues were verified through quantitative real-time polymerase chain reaction (RT-qPCR). Enrichment pathway analyses were through Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to predict the potential functions of m5C in BCBM. RESULTS The MeRIP-seq analysis identified 23,934 m5C peaks in BCBM and 21,236 m5C in BC. A total of 9,480 annotated genes (BCBM) and 8,481 annotated genes (BC) were mapped. Notably, 1,819 methylation sites in lncRNA were upregulated in BCBM, whereas 2,415 methylation sites were upregulated in BC. Significant m5C hypermethylated lncRNAs included ENST00000477316, ENST00000478098 and uc002gtt.1, whereas hypomethylated lncRNAs included ENST00000600912, ENST00000493668, ENST00000544651 and ENST00000464989. These results were verified by qPCR and MeRIP-qPCR in BC and BCBM. Considering the strong association between m5C RNA methylation regulators and lncRNA, we examined the expression levels of 13 m5C RNA methylation regulators and observed significant differences between BC tissues and adjacent normal tissues. In addition, the interaction between regulators of altered expression and the differentially expressed genes in vitro was analyzed. The GO and KEGG pathways analyses revealed that genes significantly associated with m5C sites in lncRNA were linked to the BCBM signaling pathways. CONCLUSION This uncovered significant variations in the levels and distribution of m5C in BCBM compared to BC. The findings provide a new theoretical understanding of the mechanisms of BCBM.
Collapse
Affiliation(s)
- Song Wang
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Jianran Guo
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Xinmiao Xian
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Min Li
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Anqi Zhang
- Department of Central Laboratory, Liaocheng People's Hospital, 67 Dongchang west Road, Liaocheng, 252000, Shandong, P. R. China
| | - Yujiao Liu
- Department of Stomatology, Liaocheng People's Hospital, Liaocheng, P. R. China
| | - Yifei Zhang
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Shen Chen
- Department of Breast and Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, P. R. China
| | - Guohao Gu
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Xuehua Zhang
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Dong Yan
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China
| | - Meng An
- Department of Clinical Laboratory, Liaocheng People's Hospital, 67 Dongchang west Road, Liaocheng, Shandong, 252000, P. R. China.
| | - Li Pan
- Department of Central Laboratory, Liaocheng People's Hospital, 67 Dongchang west Road, Liaocheng, 252000, Shandong, P. R. China.
| | - Bo Fu
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Liaocheng, P. R. China.
- Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, P. R. China.
| |
Collapse
|
28
|
Yang J, Zhang D, Jiang W. Long noncoding RNA as an emerging regulator of endoderm differentiation: progress and perspectives. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:11. [PMID: 40133743 PMCID: PMC11937447 DOI: 10.1186/s13619-025-00230-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/09/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
Accumulated studies have demonstrated that long noncoding RNAs (lncRNAs) play crucial regulatory roles in diverse biological processes, such as embryonic development and cell differentiation. Comprehensive transcriptome analysis identifies extensive lncRNAs, gradually elucidating their functions across various contexts. Recent studies have highlighted the essential role of lncRNAs in definitive endoderm differentiation, underscoring their importance in early development. In this review, we have analyzed the features of overlapping, proximal, and desert lncRNAs, classified by genomic location, in pluripotent stem cells (PSCs) and the differentiation derivatives. Furthermore, we focus on the endoderm lineage and review the latest advancements in lncRNA identification and their distinct regulatory mechanisms. By consolidating current knowledge, we aim to provide a clearer perspective on how lncRNAs contribute to endoderm differentiation in different manners.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
29
|
Gadad SS, Camacho CV, Gong X, Thornton M, Malladi VS, Nagari A, Sundaresan A, Nandu T, Koul S, Peng Y, Kraus WL. X-Linked Cancer-Associated Polypeptide (XCP) from lncRNA1456 Cooperates with PHF8 to Regulate Gene Expression and Cellular Pathways in Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644649. [PMID: 40196671 PMCID: PMC11974697 DOI: 10.1101/2025.03.21.644649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Recent studies have demonstrated that a subset of long "noncoding" RNAs (lncRNAs) produce functional polypeptides and proteins. In this study, we discovered a 132 amino acid protein in human breast cancer cells named XCP (X-linked Cancer-associated Polypeptide), which is encoded by lncRNA1456 (a.k.a. RHOXF1P3), a transcript previously thought to be noncoding. lncRNA1456 is a pancreas- and testis-specific RNA whose gene is located on chromosome X. We found that the expression of lncRNA1456 and XCP are highly upregulated in the luminal A, luminal B, and HER2 molecular subtypes of breast cancer. XCP modulates both estrogen-dependent and estrogen-independent growth of breast cancer cells by regulating cancer pathways, as shown in cell and xenograft models. XCP shares some homology with homeodomain-containing proteins and interacts with the histone demethylase plant homeodomain finger protein 8 (PHF8), which is also encoded by an X-linked gene. Mechanistically, XCP stimulates the histone demethylase activity of PHF8 to regulate gene expression in breast cancer cells. These findings identify XCP as a coregulator of transcription and emphasize the need to interrogate the potential functional roles of open reading frames originating from noncoding RNAs.
Collapse
Affiliation(s)
- Shrikanth S. Gadad
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Current address: Center of Emphasis in Cancer, Paul L. Foster School of Medicine, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
- These authors contributed equally to this work
| | - Cristel V. Camacho
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- These authors contributed equally to this work
| | - Xuan Gong
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Current address: Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Micah Thornton
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Computational Core Facility, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Venkat S. Malladi
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Computational Core Facility, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anusha Nagari
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Computational Core Facility, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Aishwarya Sundaresan
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Computational Core Facility, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tulip Nandu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Computational Core Facility, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sneh Koul
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Computational Core Facility, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yan Peng
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - W. Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
30
|
Wei DP, Jiang WW, Chen CX, Chen ZY, Zhou FQ, Zhang Y, Lu J. Identification and validation of autophagy-related genes in sepsis based on bioinformatics studies. Virol J 2025; 22:81. [PMID: 40114170 PMCID: PMC11924728 DOI: 10.1186/s12985-025-02683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 02/25/2025] [Indexed: 03/22/2025] Open
Abstract
We identified 14 key genes associated with mitochondrial autophagy in sepsis through differential analysis of the dataset and then analysed the identified genes for functional enrichment. The analysis of key genes and deeper analysis of key genes by molecular typing, Weighted Gene Correlation Network Analysis (WGCNA) and ceRNA were also carried out. We have also validated these key genes with clinical data. Finally, sepsis diagnostic models are constructed by combining key genes with machine learning methods. In addition, we discuss the importance of the immune system in sepsis and its relationship with signature genes, which opens up new directions for studying the role of the immune system in sepsis. Overall, our study adds new ideas to the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Dong-Po Wei
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
| | - Wei-Wei Jiang
- Department of Emergency and Critical Care Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chang-Xing Chen
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
| | - Zi-Yang Chen
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
| | - Fang-Qing Zhou
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China
| | - Yu Zhang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China.
| | - Jian Lu
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China.
- Department of Critical Care Medicine, Shanghai United Family Hospital, Shanghai, China.
| |
Collapse
|
31
|
Saxena T, Quan A, Chan E, Kozlova N, Matai L, Lee JD, Rupaimoole R, Beca F, Muranen T, Slack FJ. EGFR-induced lncRNA TRIDENT promotes drug resistance in non-small cell lung cancer via phospho-TRIM28-mediated DNA damage repair. Proc Natl Acad Sci U S A 2025; 122:e2415389122. [PMID: 40030013 PMCID: PMC11912419 DOI: 10.1073/pnas.2415389122] [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: 07/31/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) play numerous roles in cellular biology and alterations in lncRNA expression profiles have been implicated in a variety of cancers. Here, we identify and characterize a lncRNA, TRIM28 Interacting DNA damage repair Enhancing Noncoding Transcript (TRIDENT), whose expression is induced upon epithelial growth factor receptor (EGFR) activation, and which exerts pro-oncogenic functions in EGFR-driven non-small cell lung cancer. Knocking down TRIDENT leads to decreased tumor-cell proliferation in both in vitro and in vivo model systems and induces sensitization to chemotherapeutic drugs. Using ChIRP-MS analysis we identified TRIM28 as a protein interactor of TRIDENT. TRIDENT promotes phosphorylation of TRIM28 and knocking down TRIDENT leads to accumulation of DNA damage in cancer cells via decreased TRIM28 phosphorylation. Altogether, our results reveal a molecular pathway in which TRIDENT regulates TRIM28 phosphorylation to promote tumor cell growth and drug resistance. Our findings suggest that TRIDENT can be developed as a biomarker or therapeutic target for EGFR mutant non-small cell lung cancer.
Collapse
Affiliation(s)
- Tanvi Saxena
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Anan Quan
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Erica Chan
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Nina Kozlova
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Latika Matai
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Jonathan D. Lee
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Rajesha Rupaimoole
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Francisco Beca
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Taru Muranen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Frank J. Slack
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| |
Collapse
|
32
|
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 2025; 83:415-427. [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] [MESH Headings] [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
|
33
|
Li J, Ren D, Meng X, He Y, Wang L, Sheng X, Wang L. Transcriptomics-based investigation of resistance differences to swine fever between large white pigs and min pigs. Virus Res 2025; 353:199536. [PMID: 39875015 PMCID: PMC11841072 DOI: 10.1016/j.virusres.2025.199536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/25/2024] [Accepted: 01/26/2025] [Indexed: 01/30/2025]
Abstract
The genetic foundations underlying the observed disease resistance in certain indigenous pig breeds, notably the Min pigs of China, present a compelling underexplored subject of study. Exploring the mechanisms of disease resistance in these breeds could lay the groundwork for genetic improvements in pig immunity, potentially augmenting overall pig productivity. In this study, whole blood samples were collected from pre- and post- swine fever vaccinated Min and Large White pigs for transcriptome sequencing. The mRNA and lncRNA in both pig breeds were analyzed, and intra-group and inter-group comparisons were also conducted. The results indicated that a greater number of immune-related pathways such as the JAK-STAT and PI3K-AKT signaling were enriched in Min pigs. Furthermore, genes involved in inflammation and antiviral responses, including IL16, IL27, USP18, and DHX58, were upregulated in post-vaccination Min pigs compared to post-vaccination Large White pigs. This heightened immune responsiveness could contribute to the observed differences in disease resistance between Min pigs and Large White pigs.
Collapse
Affiliation(s)
- Jia Li
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China; Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Deming Ren
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiangxu Meng
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Yiyun He
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lixian Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xihui Sheng
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China.
| | - Ligang Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
34
|
Zhu J, Jian Z, Liu F, Le L. The emerging landscape of small nucleolar RNA host gene 10 in cancer mechanistic insights and clinical relevance. Cell Signal 2025; 127:111590. [PMID: 39798772 DOI: 10.1016/j.cellsig.2025.111590] [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: 09/18/2024] [Revised: 12/14/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
Small nucleolar RNA host gene 10 (SNHG10) is a newly recognized long non-coding RNA (lncRNA) with significant implications in cancer biology. Abnormal expression of SNHG10 has been observed in various solid tumors and hematological malignancies. Research conducted in vivo and in vitro has revealed that SNHG10 plays a pivotal role in numerous biological processes, including cell proliferation, apoptosis, invasion and migration, drug resistance, energy metabolism, immune evasion, as well as tumor growth and metastasis. SNHG10 regulates tumor development through several mechanisms, such as competing with microRNA (miRNA) for binding sites, modulating various signaling pathways, influencing transcriptional activity, and affecting epigenetic regulation. The diverse biological functions and intricate mechanisms of SNHG10 highlight its considerable clinical relevance, positioning it as a potential pan-cancer biomarker and therapeutic target. This review aims to summarize the role of SNHG10 in tumorigenesis and cancer progression, clarify the molecular mechanisms at play, and explore its clinical significance in cancer diagnosis and prognosis prediction, along with its therapeutic potential.
Collapse
Affiliation(s)
- Jingyu Zhu
- Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, China
| | - Zihao Jian
- Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, China
| | - Fangteng Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China.
| | - Lulu Le
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China.
| |
Collapse
|
35
|
Liu J, Yao L, Yang Y, Ma J, You R, Yu Z, Du P. A novel stemness-related lncRNA signature predicts prognosis, immune infiltration and drug sensitivity of clear cell renal cell carcinoma. J Transl Med 2025; 23:238. [PMID: 40016772 PMCID: PMC11869577 DOI: 10.1186/s12967-025-06251-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a prevalent urogenital malignancy characterized by heterogeneous patterns. Stemness is a pivotal factor in tumor progression, recurrence, and metastasis. Nevertheless, the impact of stemness-related long non-coding RNAs (SRlncRNAs) on the prognosis of ccRCC remains elusive. In this study, we aimed to delve into the SRlncRNAs of ccRCC and develop a signature for risk stratification and prognosis prediction. METHOD Gene-expression and clinical data were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. We calculated RNA stemness scores (RNAss) for the samples to evaluate their stemness. SRlncRNAs and stemness-related mRNAs (SRmRNAs) in ccRCC were identified through weighted correlation network analysis (WGCNA), which employed sophisticated statistical methodologies to identify interconnected modules of related genes. Enrichment analysis was performed to explore the potential functions of SRmRNAs. Multiple machine learning algorithms were employed to construct a prognostic signature. Samples from TCGA-KIRC and GSE29609 cohorts were designated as the training and validation cohorts, respectively. Based on their risk scores, samples were stratified into low- and high-risk groups. Prognosis analysis, immune infiltration assessment, drug sensitivity prediction, mutation landscape, and gene set enrichment analysis (GSEA) were conducted to investigate the distinct characteristics of the low- and high-risk groups. Additionally, a web-based calculator was developed to facilitate clinical application. Expression and effects of SRlncRNAs in ccRCC were further corroborated through the utilization of single-cell RNA-seq (scRNA-seq), as well as in vitro and in vivo experiments. RESULTS SRlncRNAs and SRmRNAs were identified based on RNAss and WGCNA. The least absolute shrinkage and selection operator (LASSO) in combination with multivariate Cox regression was selected as the optimal approach. Six SRlncRNAs were used to construct the prognostic signature. Samples in the low- and high-risk groups exhibited distinct characteristics in terms of prognosis, GSEA pathways, immune infiltration profiles, drug sensitivity, and mutation status. A nomogram and a web-based calculator were developed to facilitate the clinical application of the model. ScRNA-seq and RT-qPCR demonstrated the differential expression of SRlncRNAs between ccRCC tumors and normal tissues. In vitro and in vivo experiments demonstrated that downregulation of EMX2OS and LINC00944 affected the proliferation, migration, invasion, apoptosis, and metastasis of ccRCC cells. CONCLUSION We uncovered the crucial associations between SRlncRNAs and the prognosis of ccRCC. By leveraging these findings, we developed a novel SRlncRNA-related signature and a user-friendly web calculator. This signature holds great potential in facilitating risk stratification and guiding tailored treatment strategies for ccRCC patients. Both in vitro and in vivo experiments confirmed the role of SRlncRNAs in the progression of ccRCC.
Collapse
Affiliation(s)
- Jia Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lin Yao
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Yong Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jinchao Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ruijian You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ziyi Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Peng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
36
|
Wang F, Mu HF, Wang C, Tang Y, Si MY, Peng J. LncRNA PCAT6 promotes progression and metastasis of colonic neuroendocrine carcinoma via MAPK pathway. World J Gastrointest Oncol 2025; 17:96230. [PMID: 39958556 PMCID: PMC11755991 DOI: 10.4251/wjgo.v17.i2.96230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 09/18/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Colonic neuroendocrine carcinomas (NECs) are highly malignant and invasive with poor prognosis. Long noncoding RNAs (LncRNAs) participate in the tumorigenesis and metastasis of multiple cancers. AIM To detect the roles and mechanisms of lncRNA prostate cancer associated transcript 6 (PCAT6) in the progression of colonic NEC. METHODS Human NEC and adjacent normal samples were collected for immunohistochemistry staining of CgA and real-time quantitative polymerase chain reaction (RT-qPCR) of PCAT6 mRNA level. Subcutaneous xenograft tumor model and lung metastasis model were established in nude mice. The lung tissues were stained by hematoxylin and eosin to assess pulmonary metastasis. The expression of epithelial-mesenchymal transition (EMT)-related markers and pathway-related genes was measured by RT-qPCR and western blotting. CD56 expression was assessed by immunofluorescence staining. The biological functions of PCAT6 were examined by cell counting kit-8, colony formation assays, Transwell assays and wound healing assays. The interaction between PCAT6 and its potential downstream target was verified by luciferase reporter assays. RESULTS LncRNA PCAT6 was upregulated in human NEC samples and LCC-18 cells, and its high expression was positively correlated with poor prognosis in patients with colonic NEC. Additionally, the expression of PCAT6 was positively associated with the proliferation, migration, invasion, and EMT of LCC-18 cells. Moreover, PCAT6 facilitated tumor growth, lung metastasis and EMT in xenografts. Mechanistically, PCAT6 promoted the activation of MAPK to enhance the EMT in colonic NEC by targeting miR-326. CONCLUSION In conclusion, lncRNA PCAT6 accelerates the process of colonic NEC by activating ERK/p38 MAPK signaling through targeting miR-326. These results might provide useful information for exploring the potential therapeutic targets in colonic NEC.
Collapse
Affiliation(s)
- Fei Wang
- Department of General Surgery, Nanjing Tongren Hospital, Nanjing 211100, Jiangsu Province, China
| | - Hai-Feng Mu
- Department of General Surgery, Nanjing Tongren Hospital, Nanjing 211100, Jiangsu Province, China
| | - Chun Wang
- Department of General Surgery, Nanjing Tongren Hospital, Nanjing 211100, Jiangsu Province, China
| | - Yue Tang
- Department of General Surgery, Nanjing Tongren Hospital, Nanjing 211100, Jiangsu Province, China
| | - Ming-Yuan Si
- Department of Pathology, Nanjing Tongren Hospital, Nanjing 211100, Jiangsu Province, China
| | - Jing Peng
- Department of General Surgery, Nanjing Tongren Hospital, Nanjing 211100, Jiangsu Province, China
| |
Collapse
|
37
|
Saeed BI, Kumar A, Oghenemaro EF, Almutairi LA, M RM, Kumawat R, Uthirapathy S, Hulail HM, Sharma S, Ravi Kumar M. Interactions between lncRNAs and cyclins/CDKs complexes; key players in determining cancer cell response to CDKs inhibitors. Exp Cell Res 2025; 445:114406. [PMID: 39761840 DOI: 10.1016/j.yexcr.2025.114406] [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: 11/26/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/28/2025]
Abstract
Transcription takes place over a significant portion of the human genome. However, only a small portion of the transcriptome, roughly 1.2 %, consists of RNAs translated into proteins; the majority of transcripts, on the other hand, comprise a variety of RNA families with varying sizes and functions. A substantial portion of this diverse RNA universe consists of sequences longer than 200 bases, called the long non-coding RNA (lncRNA). The control of gene transcription, changes to DNA topology, nucleosome organization and structure, paraspeckle creation, and assistance for developing cellular organelles are only a few of the numerous tasks performed by lncRNA. The main focus of this study is on the function of lncRNA in controlling the levels and actions of cyclin-dependent kinase inhibitors (CDKIs). The enzymes required for the mitotic cycle's regulated progression are called cyclin-dependent kinases (CDKs). They have many degrees of regulation over their activities and interact with CDKIs as their crucial mechanisms. Interestingly, culminating evidence has clarified that lncRNAs are associated with several illnesses and use CDKI regulation to control cellular function. Nonetheless, despite the abundance of solid evidence in the literature, it still seems unlikely that lncRNA will have much of an impact on controlling cell proliferation or modulating CDKIs.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq.
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named After the First President of Russia Boris Yeltsin, Ekaterinburg, 620002, Russia; Department of Mechanical Engineering, Karpagam Academy of Higher Education, Coimbatore, 641021, India.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, PMB 1, Abraka, Delta State, Nigeria.
| | - Layla A Almutairi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia.
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India.
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Subasini Uthirapathy
- Faculty of Pharmacy, Pharmacology Department, Tishk International University, Erbil, Kurdistan Region of Iraq, Iraq.
| | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq.
| | - Shilpa Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India.
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India.
| |
Collapse
|
38
|
John A, Almulla N, Elboughdiri N, Gacem A, Yadav KK, Abass AM, Alam MW, Wani AW, Bashir SM, Rab SO, Kumar A, Wani AK. Non-coding RNAs in Cancer: Mechanistic insights and therapeutic implications. Pathol Res Pract 2025; 266:155745. [PMID: 39637712 DOI: 10.1016/j.prp.2024.155745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Non-coding RNAs have gathered significant attention for their unique roles in biological regulation. Across a broad spectrum of developmental processes and diseases, particularly in human malignancies, ncRNAs play pivotal roles in regulatory mechanisms. MicroRNAs, long noncoding RNAs, and small nucleolar RNAs stand out among the diverse forms of ncRNAs that have been implicated in cancer. MiRNAs, classified as short non-coding RNAs, modulate gene expression by binding to messenger RNA molecules, thereby inhibiting their translation. Altered miRNA expression has been associated with the onset and progression of various malignancies, including lung, breast, and prostate cancer. In contrast, lncRNAs, characterized as longer ncRNAs, exert control over gene expression through various mechanisms, such as chromatin remodelling and gene silencing. This review offers a comprehensive examination of the numerous ncRNAs that have emerged as crucial regulators of gene expression, playing implicated roles in the initiation and progression of diverse cancers.
Collapse
Affiliation(s)
- Arjumand John
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Punjab 144411, India
| | - Nuha Almulla
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Noureddine Elboughdiri
- Chemical Engineering Department, College of Engineering, University of Ha'il, P.O. Box 2440, Ha'il 81441, Saudi Arabia
| | - Amel Gacem
- Department of Physics, Faculty of Sciences, University 20 Aout, Skikda 1955, Algeria
| | - Krishna Kumar Yadav
- Department of VLSI Microelectronics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai - 602105, Tamil Nadu, India; Environmental and Atmospheric Sciences Research Group, Scientific Research Center, Al-Ayen University, Thi-Qar, Nasiriyah 64001, Iraq
| | - Anass M Abass
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Mir Waqas Alam
- Department of Physics, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia.
| | - Ab Waheed Wani
- Department of Horticulture, School of Agriculture, Lovely Professional University, Jalandhar, Punjab 144411, India
| | - Showkeen Muzamil Bashir
- Biochemistry & Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir 190006, India
| | - Safia Obaidur Rab
- Central Labs, King Khalid University, AlQura'a, P.O. Box 960, Abha, Saudi Arabia; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg 620002, Russia; Department of Technical Sciences, Western Caspian University, Baku, Azerbaijan; Department of Mechanical Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Punjab 144411, India.
| |
Collapse
|
39
|
Saadh MJ, Menon SV, Verma R, Siva Prasad GV, Allela OQB, Mahdi MS, Ahmad N, Husseen B. LncRNA CRNDE and HOTAIR: Molecules behind the scenes in the progression of gastrointestinal cancers through regulating microRNAs. Pathol Res Pract 2025; 266:155778. [PMID: 39721094 DOI: 10.1016/j.prp.2024.155778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Gastrointestinal (GI) cancers, such as gastric cancer, hepatocellular carcinoma, colorectal cancer, and esophageal cancer, pose a significant medical and economic burden globally, accounting for the majority of new cancer cases and deaths each year. A lack of knowledge about the molecular mechanisms of GI cancers is reflected in the low efficacy of treatment for individuals with late stage and recurring illness. Understanding the molecular pathways that promote the growth of GI cancers may open doors for their therapy. Numerous long non-coding RNAs (lncRNAs) that are produced differently in normal and malignant tissues have been discovered by genome-wide techniques. The role of lncRNAs in the diagnosis, proliferation, metastasis, and drug resistance of different GI cancers has been investigated in recent research. LncRNAs may affect transcription, epigenetic modifications, protein/RNA stability, translation, and post-translational modifications via their interactions with DNA, RNAs, and proteins. Also, by functioning as competing endogenous RNAs (ceRNAs), they control the synthesis of certain microRNAs (miRNAs), which in turn modify the downstream target molecules of these miRNAs. Based on recent studies, lncRNAs in particular, CRNDE and HOTAIR, sponge different miRNAs and their downstream genes, which in turn regulate GI cancers development, including cell proliferation, invasion, migration, and chemoresistance. In this comprehensive review, we present an overview of the biological roles of CRNDE and HOTAIR and their associated mechanisms, miRNAs/mRNA pathways, in various GI cancers, encompassing colorectal cancer, hepatocellular carcinoma, esophageal cancer, and gastric cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Sahibzada Ajit Singh Nagar, Jhanjeri, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | | | | | - Nabeel Ahmad
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh-247341, India; Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand- 831001, India.
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
40
|
Zhu X, Zhong M, Wang Q, Zhang M. LncRNA MIR600HG inhibits laryngeal cancer development by mediating the miR-424-5p/BTG2 axis. Cancer Sci 2025; 116:544-558. [PMID: 39618050 PMCID: PMC11786319 DOI: 10.1111/cas.16404] [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: 07/26/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 02/02/2025] Open
Abstract
Laryngeal carcinoma is the predominant kind of tumor seen under the category of head and neck malignancies. LncRNA MIR600HG affects tumor morphology in numerous cancer types. However, the function of MIR600HG in laryngeal cancer remains unclear. Protein and gene expressions were analyzed by using western blot and quantitative real time polymerase chain reaction. Cells proliferation and migration were evaluated by EdU and transwell assays. Flow cytometry was performed to detect cells apoptosis. The interaction between MIR600HG or B-cell translocation gene 2 (BTG2) and miR-424-5p was analyzed by dual luciferase reporter assay and RNA immunoprecipitation. The expression of MIR600HG in laryngeal cancer tissues was lower than that in normal tissues, and low expression of MIR600HG was associated with poor prognosis in laryngeal cancer. Furthermore, overexpression of MIR600HG resulted in a reduction in cellular proliferation and the promotion of apoptosis in both HEp-2 and Tu-212. Mechanically, miR-424-5p was a direct target of MIR600HG, and overexpression of MIR600HG reduced miR-424-5p expression. Furthermore, BTG2 was a target gene of miR-424-5p and miR-424-5p upregulation suppressed the expression of BTG2. In addition, overexpression of BTG2 inhibited laryngeal cancer progression, whereas MIR600HG knockdown or miR-424-5p overexpression reversed the role of BTG2. This work suggested that MIR600HG represses laryngeal tumor development by regulating the miR-424-5p/BTG2 axis, which provides new molecules for early diagnosis of laryngeal cancer in the future.
Collapse
Affiliation(s)
- Xiaowen Zhu
- General surgery fourth wardThe First Affiliated Hospital of Jiamusi UniversityJiamusiHeilongjiangChina
| | - Min Zhong
- General surgery fourth wardThe First Affiliated Hospital of Jiamusi UniversityJiamusiHeilongjiangChina
| | - Qingdong Wang
- Department of AnesthesiologyThe First Affiliated Hospital of Jiamusi UniversityJiamusiHeilongjiangChina
| | - MeiJia Zhang
- Department of OtolaryngologyThe First Affiliated Hospital of Jiamusi UniversityJiamusiHeilongjiangChina
| |
Collapse
|
41
|
Niinuma T, Kitajima H, Sato T, Ogawa T, Ishiguro K, Kai M, Yamamoto E, Hatanaka Y, Nojima I, Toyota M, Yorozu A, Sekiguchi S, Tohse N, Furuhashi M, Ohguro H, Miyazaki A, Suzuki H. LINC02154 promotes cell cycle and mitochondrial function in oral squamous cell carcinoma. Cancer Sci 2025; 116:393-405. [PMID: 39576738 PMCID: PMC11786299 DOI: 10.1111/cas.16379] [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: 06/30/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 11/24/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) play pivotal roles in the development of human malignancies, though their involvement in oral squamous cell carcinoma (OSCC) remains incompletely understood. Using The Cancer Genome Atlas (TCGA) dataset, we analyzed expression of 7840 lncRNAs in primary head and neck squamous cell carcinoma (HNSCC) and found that upregulation of LINC02154 is associated with a poorer prognosis. LINC02154 knockdown in OSCC cell lines induced cell cycle arrest and apoptosis, and significantly attenuated tumor growth in vitro and in vivo. Notably, depletion of LINC02154 downregulated FOXM1, a master regulator of cell cycle-related genes. RNA pulldown and mass spectrometry analyses identified a series of proteins that could potentially interact with LINC02154, including HNRNPK and LRPPRC. HNRNPK stabilizes FOXM1 expression by interacting with the 3'-UTR of FOXM1 mRNA, which suggests LINC02154 and HNRNPK promote cell cycling by regulating FOXM1 expression. Additionally, LINC02154 positively regulates HNRNPK expression by inhibiting microRNAs targeting HNRPNK. Moreover, LINC02154 affects mitochondrial function by interacting with LRPPRC. Depletion of LINC02154 suppressed expression of mitochondrial genes, including MTCO1 and MTCO2, and inhibited mitochondrial respiratory function in OSCC cells. These results suggest that LINC02154 exerts its oncogenic effects by modulating the cell cycle and oxidative phosphorylation in OSCC, highlighting LINC02154 as a potential therapeutic target.
Collapse
Affiliation(s)
- Takeshi Niinuma
- Department of Molecular BiologySapporo Medical University School of MedicineSapporoJapan
| | - Hiroshi Kitajima
- Department of Molecular BiologySapporo Medical University School of MedicineSapporoJapan
| | - Tatsuya Sato
- Department of Cellular Physiology and Signal TransductionSapporo Medical University School of MedicineSapporoJapan
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Toshifumi Ogawa
- Department of Cellular Physiology and Signal TransductionSapporo Medical University School of MedicineSapporoJapan
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Kazuya Ishiguro
- Department of Molecular BiologySapporo Medical University School of MedicineSapporoJapan
| | - Masahiro Kai
- Department of Molecular BiologySapporo Medical University School of MedicineSapporoJapan
| | - Eiichiro Yamamoto
- Department of Molecular BiologySapporo Medical University School of MedicineSapporoJapan
| | - Yui Hatanaka
- Department of Oral SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Iyori Nojima
- Division of Cell Bank, Biomedical Research, Education and Instrumentation CenterSapporo Medical University School of MedicineSapporoJapan
| | - Mutsumi Toyota
- Department of Molecular BiologySapporo Medical University School of MedicineSapporoJapan
| | - Akira Yorozu
- Department of Otolaryngology – Head and Neck SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Shohei Sekiguchi
- Department of Oral SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Noritsugu Tohse
- Department of Cellular Physiology and Signal TransductionSapporo Medical University School of MedicineSapporoJapan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Hiroshi Ohguro
- Department of OphthalmologySapporo Medical University School of MedicineSapporoJapan
| | - Akihiro Miyazaki
- Department of Oral SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Hiromu Suzuki
- Department of Molecular BiologySapporo Medical University School of MedicineSapporoJapan
| |
Collapse
|
42
|
Sethi SC, Singh R, Sahay O, Barik GK, Kalita B. Unveiling the hidden gem: A review of long non-coding RNA NBAT-1 as an emerging tumor suppressor and prognostic biomarker in cancer. Cell Signal 2025; 126:111525. [PMID: 39592019 DOI: 10.1016/j.cellsig.2024.111525] [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: 08/20/2024] [Revised: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
Previously considered junk or non-functional, long non-coding RNAs (lncRNAs) have emerged over the past few decades as pivotal components in both physiological and pathological processes, including cancer. Neuroblastoma-associated transcript-1 (NBAT-1) was initially discovered a decade ago as a risk-associated tumor suppressor lncRNA in neuroblastoma (NB). Subsequent studies have consistently demonstrated that NBAT-1 serves as a dedicated tumor suppressor in many cancers. NBAT-1 is significantly downregulated in cancer, which is closely linked to higher histological grades, increased metastasis, and poor survival in cancer patients suggesting NBAT-1's potential as a prognostic biomarker. In this review, we delve into the current body of literature, elucidating the tumor-suppressive roles of NBAT-1 and the underlying regulatory mechanisms in the context of human malignancies. Additionally, we shed light on the mechanisms contributing to the diminished expression of NBAT-1 and its potential as both a prognostic biomarker and a promising therapeutic target in cancer.
Collapse
Affiliation(s)
- Subhash Chandra Sethi
- Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ragini Singh
- Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Osheen Sahay
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ganesh Kumar Barik
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Bhargab Kalita
- Amrita Research Center, Amrita Vishwa Vidyapeetham, Amrita Hospital, Mata Amritanandamayi Marg, Faridabad 121002, India.
| |
Collapse
|
43
|
Gugnoni M, Kashyap MK, Wary KK, Ciarrocchi A. lncRNAs: the unexpected link between protein synthesis and cancer adaptation. Mol Cancer 2025; 24:38. [PMID: 39891197 PMCID: PMC11783725 DOI: 10.1186/s12943-025-02236-7] [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: 11/19/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
Cancer progression relies on the ability of cells to adapt to challenging environments overcoming stresses and growth constraints. Such adaptation is a multifactorial process that depends on the rapid reorganization of many basic cellular mechanisms. Protein synthesis is often dysregulated in cancer, and translational reprogramming is emerging as a driving force of cancer adaptive plasticity. Long non-coding RNAs (lncRNAs) represent the main product of genome transcription. They outnumber mRNAs by an order of magnitude and their expression is regulated in an extremely specific manner depending on context, space and time. This heterogeneity is functional and allows lncRNAs to act as context-specific, fine-tuning controllers of gene expression. Multiple recent evidence underlines how, besides their consolidated role in transcription, lncRNAs are major players in translation control. Their capacity to establish multiple and highly dynamic interactions with proteins and other transcripts makes these molecules able to play a central role across all phases of protein synthesis. Even if through a myriad of different mechanisms, the action of these transcripts is dual. On one hand, by modulating the overall translation speed, lncRNAs participate in the process of metabolic adaptation of cancer cells under stress conditions. On the other hand, by prioritizing the synthesis of specific transcripts they help cancer cells to maintain high levels of essential oncogenes. In this review, we aim to discuss the most relevant evidence regarding the involvement of lncRNAs in translation regulation and to discuss how this specific function may affect cancer plasticity and resistance to stress. We also expect to provide one of the first collective perspectives on the way these transcripts modulate gene expression beyond transcription.
Collapse
Affiliation(s)
- Mila Gugnoni
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| | - Kishore K Wary
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago, IL, USA.
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
44
|
Hamamoto K, Zhu G, Lai Q, Lesperance J, Luo H, Li Y, Nigam N, Sharma A, Yang FC, Claxton D, Qiu Y, Aplan PD, Xu M, Huang S. HoxBlinc lncRNA reprograms CTCF-independent TADs to drive leukemic transcription and HSC dysregulation in NUP98-rearranged leukemia. J Clin Invest 2025; 135:e184743. [PMID: 39883527 PMCID: PMC11957699 DOI: 10.1172/jci184743] [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: 07/11/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025] Open
Abstract
Although nucleoporin 98 (NUP98) fusion oncogenes often drive aggressive pediatric leukemia by altering chromatin structure and expression of homeobox (HOX) genes, underlying mechanisms remain elusive. Here, we report that the Hoxb-associated lncRNA HoxBlinc was aberrantly activated in NUP98-PHF23 fusion-driven leukemias. HoxBlinc chromatin occupancies led to elevated mixed-lineage leukemia 1 (MLL1) recruitment and aberrant homeotic topologically associated domains (TADs) that enhanced chromatin accessibilities and activated homeotic/hematopoietic oncogenes. HoxBlinc depletion in NUP98 fusion-driven leukemia impaired HoxBlinc binding, TAD integrity, MLL1 recruitment, and the MLL1-driven chromatin signature within HoxBlinc-defined TADs in a CCCTC-binding factor-independent (CTCF-independent) manner, leading to inhibited homeotic/leukemic oncogenes that mitigated NUP98 fusion-driven leukemogenesis in xenografted mouse models. Mechanistically, HoxBlinc overexpression in the mouse hematopoietic compartment induced leukemias resembling those in NUP98-PHF23-knockin (KI) mice via enhancement of HoxBlinc chromatin binding, TAD formation, and Hox gene aberration, leading to expansion of hematopoietic stem and progenitor cell and myeloid/lymphoid cell subpopulations. Thus, our studies reveal a CTCF-independent role of HoxBlinc in leukemic TAD organization and oncogene-regulatory networks.
Collapse
Affiliation(s)
- Karina Hamamoto
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ganqian Zhu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Qian Lai
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Julia Lesperance
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Huacheng Luo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ying Li
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Nupur Nigam
- Genetics Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Arati Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Feng-Chun Yang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David Claxton
- Division of Hematology/Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
- Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Yi Qiu
- Penn State Cancer Institute, Hershey, Pennsylvania, USA
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Peter D. Aplan
- Genetics Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Mingjiang Xu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Suming Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
- Penn State Cancer Institute, Hershey, Pennsylvania, USA
| |
Collapse
|
45
|
Gong L, Zou C, Zhang H, Yang F, Qi G, Ma Z. Landscape of Noncoding RNA in the Hypoxic Tumor Microenvironment. Genes (Basel) 2025; 16:140. [PMID: 40004471 PMCID: PMC11855738 DOI: 10.3390/genes16020140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Amidst the prevalent and notable characteristic of a hypoxic microenvironment present in the majority of solid tumors, a burgeoning number of studies have revealed the significance of noncoding RNAs (ncRNAs) in hypoxic tumor regions. The transcriptome of cancers is highly heterogeneous, with noncoding transcripts playing crucial roles. Long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) are two distinctive classes of ncRNA that are garnering increasing attention. Biologically, they possess intriguing properties and possess significant regulatory functions. Clinically, they present as promising biomarkers and therapeutic targets. Additionally, recent research has evaluated the clinical applications of these ncRNAs in RNA-based treatments and noninvasive liquid biopsies. This review provides a comprehensive summary of recent studies on lncRNAs and circRNAs within the hypoxic tumor microenvironment. Furthermore, the clinical significance of lncRNAs and circRNAs in cancer diagnosis and treatment is emphasized, which could pave the way for the development of effective targeted therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (L.G.); (C.Z.); (H.Z.); (F.Y.); (G.Q.)
| |
Collapse
|
46
|
Yuan H, Zhou L, Hu W, Yang M. LINC00626 drives tamoxifen resistance in breast cancer cells by interaction with UPF1. Sci Rep 2025; 15:2997. [PMID: 39848992 PMCID: PMC11757752 DOI: 10.1038/s41598-025-86287-2] [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: 07/20/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025] Open
Abstract
Although tamoxifen is commonly utilized as adjuvant therapy for Estrogen Receptor alpha (ERα)-positive breast cancer patients, approximately 30-50% of individuals treated with tamoxifen experience relapse. Therefore, it is essential to investigate additional factors besides ERα that influence the estrogen response. In this study, cross-analysis of databases were performed, and the results revealed a significant association between LINC00626 and ERα signaling as well as increased expression levels of this gene in tamoxifen-resistant cells. LINC00626 is a novel ERα-regulated long non-coding RNA (lncRNA) that has not yet been examined for its potential contribution to endocrine therapy resistance. This study revealed that the upregulation of LINC00626 in breast cancer was associated with poor overall survival in patients. Additionally, ERα signaling was found to transcriptionally regulate LINC00626 expression, thereby promoting cancer progression and enhancing resistance to tamoxifen in breast cancer cells via the regulation of UPF1 expression. Depletion of LINC00626 restored sensitivity to tamoxifen by activating the PERK-ATF4-CHOP signaling pathway via UPF1. These findings support the role of LINC00626 as a potential therapeutic target for combating tamoxifen resistance.
Collapse
Affiliation(s)
- Hui Yuan
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
- The Second Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| | - Lianbang Zhou
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| | - Min Yang
- The Second Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| |
Collapse
|
47
|
Cai ZR, Zheng YQ, Hu Y, Ma MY, Wu YJ, Liu J, Yang LP, Zheng JB, Tian T, Hu PS, Liu ZX, Zhang L, Xu RH, Ju HQ. Construction of exosome non-coding RNA feature for non-invasive, early detection of gastric cancer patients by machine learning: a multi-cohort study. Gut 2025:gutjnl-2024-333522. [PMID: 39753334 DOI: 10.1136/gutjnl-2024-333522] [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: 08/01/2024] [Accepted: 12/08/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND AND OBJECTIVE Gastric cancer (GC) remains a prevalent and preventable disease, yet accurate early diagnostic methods are lacking. Exosome non-coding RNAs (ncRNAs), a type of liquid biopsy, have emerged as promising diagnostic biomarkers for various tumours. This study aimed to identify a serum exosome ncRNA feature for enhancing GC diagnosis. DESIGNS Serum exosomes from patients with GC (n=37) and healthy donors (n=20) were characterised using RNA sequencing, and potential biomarkers for GC were validated through quantitative reverse transcription PCR (qRT-PCR) in both serum exosomes and tissues. A combined diagnostic model was developed using LASSO-logistic regression based on a cohort of 518 GC patients and 460 healthy donors, and its diagnostic performance was evaluated via receiver operating characteristic curves. RESULTS RNA sequencing identified 182 candidate biomarkers for GC, of which 31 were validated as potential biomarkers by qRT-PCR. The combined diagnostic score (cd-score), derived from the expression levels of four long ncRNAs (RP11.443C10.1, CTD-2339L15.3, LINC00567 and DiGeorge syndrome critical region gene (DGCR9)), was found to surpass commonly used biomarkers, such as carcinoembryonic antigen, carbohydrate antigen 19-9 (CA19-9) and CA72-4, in distinguishing GC patients from healthy donors across training, testing and external validation cohorts, with AUC values of 0.959, 0.942 and 0.949, respectively. Additionally, the cd-score could effectively identify GC patients with negative gastrointestinal tumour biomarkers and those in early-stage. Furthermore, molecular biological assays revealed that knockdown of DGCR9 inhibited GC tumour growth. CONCLUSIONS Our proposed serum exosome ncRNA feature provides a promising liquid biopsy approach for enhancing the early diagnosis of GC.
Collapse
Affiliation(s)
- Ze-Rong Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yong-Qiang Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yan Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Meng-Yao Ma
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, People's Republic of China
| | - Yi-Jin Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jia Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Lu-Ping Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jia-Bo Zheng
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Tian Tian
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, People's Republic of China
| | - Pei-Shan Hu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Lin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People's Republic of China
| |
Collapse
|
48
|
Che R, Panah M, Mirani B, Knowles K, Ostapovich A, Majumdar D, Chen X, DeSimone J, White W, Noonan M, Luo H, Alexandrov A. Identification of Human Pathways Acting on Nuclear Non-Coding RNAs Using the Mirror Forward Genetic Approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.26.615073. [PMID: 39386709 PMCID: PMC11463631 DOI: 10.1101/2024.09.26.615073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Despite critical roles in diseases, human pathways acting on strictly nuclear non-coding RNAs have been refractory to forward genetics. To enable their forward genetic discovery, we developed a single-cell approach that "Mirrors" activities of nuclear pathways with cytoplasmic fluorescence. Application of Mirror to two nuclear pathways targeting MALAT1's 3' end, the pathway of its maturation and the other, the degradation pathway blocked by the triple-helical Element for Nuclear Expression (ENE), identified nearly all components of three complexes: Ribonuclease P and the RNA Exosome, including nuclear DIS3, EXOSC10, and C1D, as well as the Nuclear Exosome Targeting (NEXT) complex. Additionally, Mirror identified DEAD-box helicase DDX59 associated with the genetic disorder Oral-Facial-Digital syndrome (OFD), yet lacking known substrates or roles in nuclear RNA degradation. Knockout of DDX59 exhibits stabilization of the full-length MALAT1 with a stability-compromised ENE and increases levels of 3'-extended forms of small nuclear RNAs. It also exhibits extensive retention of minor introns, including in OFD-associated genes, suggesting a mechanism for DDX59 association with OFD. Mirror efficiently identifies pathways acting on strictly nuclear non-coding RNAs, including essential and indirectly-acting components, and, as a result, uncovers unexpected links to human disease.
Collapse
Affiliation(s)
- Rui Che
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
- Clemson University Center for Human Genetics, Greenwood, SC 29646, USA
| | - Monireh Panah
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
- Clemson University Center for Human Genetics, Greenwood, SC 29646, USA
| | - Bhoomi Mirani
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
- Clemson University Center for Human Genetics, Greenwood, SC 29646, USA
| | - Krista Knowles
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
- Clemson University Center for Human Genetics, Greenwood, SC 29646, USA
| | - Anastacia Ostapovich
- Dept. of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06536, USA
| | - Debarati Majumdar
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
- Clemson University Center for Human Genetics, Greenwood, SC 29646, USA
| | - Xiaotong Chen
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
| | - Joseph DeSimone
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
| | - William White
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
| | - Megan Noonan
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
| | - Hong Luo
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
| | - Andrei Alexandrov
- Dept. of Genetics and Biochemistry, Clemson University, Clemson, SC 29631, USA
- Clemson University Center for Human Genetics, Greenwood, SC 29646, USA
| |
Collapse
|
49
|
Doghish AS, Mageed SSA, Zaki MB, Abd-Elmawla MA, Sayed GA, Hatawsh A, Aborehab NM, Moussa R, Mohammed OA, Abdel-Reheim MA, Elimam H. Role of long non-coding RNAs and natural products in prostate cancer: insights into key signaling pathways. Funct Integr Genomics 2025; 25:16. [PMID: 39821470 DOI: 10.1007/s10142-025-01526-z] [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: 11/05/2024] [Revised: 12/18/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
Prostate cancer (PC) ranks among the most prevalent cancers in males. Recent studies have highlighted intricate connections between long non-coding RNAs (lncRNAs), natural products, and cellular signaling in PC development. LncRNAs, which are RNA transcripts without protein-coding function, influence cell growth, programmed cell death, metastasis, and resistance to treatments through pathways like PI3K/AKT, WNT/β-catenin, and androgen receptor signaling. Certain lncRNAs, including HOTAIR and PCA3, are associated with PC progression, with potential as diagnostic markers. Natural compounds, such as curcumin and resveratrol, demonstrate anticancer effects by targeting these pathways, reducing tumor growth, and modulating lncRNA expression. For instance, curcumin suppresses HOTAIR levels, hindering PC cell proliferation and invasion. The interaction between lncRNAs and natural compounds may open new avenues for therapy, as these substances can simultaneously impact multiple signaling pathways. These complex interactions offer promising directions for developing innovative PC treatments, enhancing diagnostics, and identifying new biomarkers for improved prevention and targeted therapy. This review aims to map the multifaceted relationship among natural products, lncRNAs, and signaling pathways in PC pathogenesis, focusing on key pathways such as AR, PI3K/AKT/mTOR, WNT/β-catenin, and MAPK, which are crucial in PC progression and therapy resistance. Regulation of these pathways by natural products and lncRNAs could lead to new insights into biomarker identification, preventive measures, and targeted PC therapies.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, CairoE, 11829, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ghadir A Sayed
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, 11829, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26th of July Corridor, Sheikh Zayed City, Giza, 12588, Egypt
| | - Nora M Aborehab
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Rewan Moussa
- School Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | | | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| |
Collapse
|
50
|
Chen JF, Ye SZ, Wang KJ, Meng XY, Yang BB, Wu KR, Ma Q. Long non-coding RNA OSTM1-AS1 promotes renal cell carcinoma progression by sponging miR-491-5p and upregulating MMP-9. Sci Rep 2025; 15:359. [PMID: 39747324 PMCID: PMC11696353 DOI: 10.1038/s41598-024-83154-4] [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: 03/31/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) have been recognized as essential regulators in various human malignancies. Hundreds of lncRNAs were known to be abnormally expressed in renal cell carcinoma (RCC) through a lncRNA expression microarray, among which lncRNA OSTM1 antisense RNA 1(OSTM1-AS1) was revealed as one of the most abundant lncRNAs. However, the function of OSTM1-AS1 in RCC remains unknown. Here, we examined OSTM1-AS1 functional roles and mechanism in RCC development. OSTM1-AS1 expression was significantly highly expressed among RCC tissue specimens and cell lines. Functionally, OSTM1-AS1 knockdown significantly suppressed cell proliferation, migration along with metastasis of RCC cells. Mechanistically, miR-491-5p was targeted via OSTM1-AS1, and down-regulation of miR-491-5p reversed OSTM1-AS1 knockdown impact on RCC migration and invasion. MMP-9 was targeted via miR-491-5p, and MMP-9 overexpression reversed miR-491-5p or OSTM1-AS1 knockdown impact on cell migration and invasion. MMP-9 abundance was decreased by OSTM1-AS1 silence, that was reduced by miR-491-5p deficiency. Importantly, our investigation revealed that OSTM1-AS1 has the ability to interact with miR-491-5p, thereby increasing the MMP-9 expression. The in vivo trial demonstrated that OSTM1-AS1 suppression resulted in tumor growth inhibition among nude mice. In summary, our findings indicate, for the first time, at least to the best of our knowledge, that OSTM1-AS1 serves as an oncogene among RCC by promoting proliferation, invasion, and metastasis through its targeting of the miR-491-5p/MMP9 axis. Therefore, this axis could represent a promising alternative therapeutic target for RCC treatment.
Collapse
Affiliation(s)
- Jun-Feng Chen
- Translational Research Laboratory for Urology, The Key Laboratory of Ningbo City, Ningbo Clinical Research Center for Urological Disease, Comprehensive Urogenital Cancer Center, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China
| | - Sha-Zhou Ye
- Translational Research Laboratory for Urology, The Key Laboratory of Ningbo City, Ningbo Clinical Research Center for Urological Disease, Comprehensive Urogenital Cancer Center, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China
| | - Ke-Jie Wang
- Translational Research Laboratory for Urology, The Key Laboratory of Ningbo City, Ningbo Clinical Research Center for Urological Disease, Comprehensive Urogenital Cancer Center, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China
| | - Xiang-Yu Meng
- Translational Research Laboratory for Urology, The Key Laboratory of Ningbo City, Ningbo Clinical Research Center for Urological Disease, Comprehensive Urogenital Cancer Center, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China
| | - Bin-Bin Yang
- Department of Urology, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China
| | - Ke Rong Wu
- Translational Research Laboratory for Urology, The Key Laboratory of Ningbo City, Ningbo Clinical Research Center for Urological Disease, Comprehensive Urogenital Cancer Center, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China.
- Department of Urology, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China.
| | - Qi Ma
- Translational Research Laboratory for Urology, The Key Laboratory of Ningbo City, Ningbo Clinical Research Center for Urological Disease, Comprehensive Urogenital Cancer Center, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China.
- Comprehensive Genitourinary Cancer Center, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo, 315010, Zhejiang, China.
- Yi-Huan Genitourinary Cancer Group, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|