1
|
Chen Q, Xu X, Li S, Xiong T. LncRNA regulation in ischemic stroke and their application prospects. Neural Regen Res 2026; 21:1058-1073. [PMID: 40145979 DOI: 10.4103/nrr.nrr-d-24-00924] [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: 08/13/2024] [Accepted: 12/07/2024] [Indexed: 03/28/2025] Open
Abstract
Ischemic stroke is a serious medical event that cannot be predicted in advance and can have long-lasting effects on patients, families, and communities. A deeper understanding of the changes in gene expression and the fundamental molecular mechanisms involved could help address this critical issue. In recent years, research into regulatory long non-coding (lnc)RNAs, a diverse group of RNA molecules with regulatory functions, has emerged as a promising direction in the study of cerebral infarction. This review paper aims to provide a comprehensive exploration of the roles of regulatory lncRNAs in cerebral infarction, as well as potential strategies for their application in clinical settings. LncRNAs have the potential to act as "sponges" that attract specific microRNAs, thereby regulating the expression of microRNA target genes. These interactions influence various aspects of ischemic stroke, including reperfusion-induced damage, cell death, immune responses, autophagy, angiogenesis, and the generation of reactive oxygen species. We highlight several regulatory lncRNAs that have been utilized in animal model treatments, including lncRNA NKILA, lncRNA Meg8, and lncRNA H19. Additionally, we discuss lncRNAs that have been used as biomarkers for the diagnosis and prognosis of cerebral infarction, such as lncRNA FOXO3, lncRNA XIST, and lncRNA RMST. The lncRNAs hold potential for genetic-level treatments in patients. However, numerous challenges, including inefficiency, low targeting accuracy, and side effects observed in preliminary studies, indicate the need for thorough investigation. The application of lncRNAs in ischemic stroke presents challenges that require careful and extensive validation.
Collapse
Affiliation(s)
- Qianqian Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Xiangyi Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Shun Li
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tianqing Xiong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, Jiangsu Province, China
| |
Collapse
|
2
|
Sun Y, Pang X, Huang X, Liu D, Huang J, Zheng P, Wei Y, Pang C. Potential mechanisms of non-coding RNA regulation in Alzheimer's disease. Neural Regen Res 2026; 21:265-280. [PMID: 39851253 PMCID: PMC12094571 DOI: 10.4103/nrr.nrr-d-24-00696] [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: 06/23/2024] [Revised: 09/02/2024] [Accepted: 10/19/2024] [Indexed: 01/26/2025] Open
Abstract
Alzheimer's disease, a progressively degenerative neurological disorder, is the most common cause of dementia in the elderly. While its precise etiology remains unclear, researchers have identified diverse pathological characteristics and molecular pathways associated with its progression. Advances in scientific research have increasingly highlighted the crucial role of non-coding RNAs in the progression of Alzheimer's disease. These non-coding RNAs regulate several biological processes critical to the advancement of the disease, offering promising potential as therapeutic targets and diagnostic biomarkers. Therefore, this review aims to investigate the underlying mechanisms of Alzheimer's disease onset, with a particular focus on microRNAs, long non-coding RNAs, and circular RNAs associated with the disease. The review elucidates the potential pathogenic processes of Alzheimer's disease and provides a detailed description of the synthesis mechanisms of the three aforementioned non-coding RNAs. It comprehensively summarizes the various non-coding RNAs that have been identified to play key regulatory roles in Alzheimer's disease, as well as how these non-coding RNAs influence the disease's progression by regulating gene expression and protein functions. For example, miR-9 targets the UBE4B gene, promoting autophagy-mediated degradation of Tau protein, thereby reducing Tau accumulation and delaying Alzheimer's disease progression. Conversely, the long non-coding RNA BACE1-AS stabilizes BACE1 mRNA, promoting the generation of amyloid-β and accelerating Alzheimer's disease development. Additionally, circular RNAs play significant roles in regulating neuroinflammatory responses. By integrating insights from these regulatory mechanisms, there is potential to discover new therapeutic targets and potential biomarkers for early detection and management of Alzheimer's disease. This review aims to enhance the understanding of the relationship between Alzheimer's disease and non-coding RNAs, potentially paving the way for early detection and novel treatment strategies.
Collapse
Affiliation(s)
- Yue Sun
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Xinping Pang
- School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu Province, China
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Dinglu Liu
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Jingyue Huang
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Pengtao Zheng
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Yanyu Wei
- National Key Laboratory of Science and Technology on Vacuum Electronics, School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Chaoyang Pang
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| |
Collapse
|
3
|
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
|
4
|
Zhao L, Huang J, Li Y, Wu S. LncRNA transcriptome analysis of rainbow trout ( Oncorhynchus mykiss) skin infected with IHNV reveals that lncRNA SARL/miR-205-z/ SOCS3 axis negatively regulates antiviral immunity mechanisms. Virulence 2025; 16:2486990. [PMID: 40287819 PMCID: PMC12036486 DOI: 10.1080/21505594.2025.2486990] [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: 11/28/2024] [Revised: 02/06/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are new gene regulators involved in various biological processes. However, the regulatory effect of lncRNA on the rainbow trout (Oncorhynchus mykiss) antiviral immune response has not been reported. Here, we measured lncRNA profiles at 48 hpi compared to the control group, expression levels of lncRNA, miRNA, and gene, and lncRNA SARL/miR-205-z/SOCS3 functions after rainbow trout skin infected with infectious haematopoietic necrosis virus (IHNV) by RNA-seq, qRT-PCR, and overexpression and inhibition assays. Transcriptome analysis identified twelve upregulated and four downregulated DElncRNAs. Twelve key immune-related competing endogenous RNA (ceRNA) networks were identified, and the target genes were enriched in the TLR, RLR, NLR, and p53 signalling pathways. Expression patterns suggested that changes in lncRNA SARL, miR-205-z, and SOCS3 expression presented a ceRNA regulatory relationship. Further studies demonstrated that the lncRNA SARL was a ceRNA of SOCS3 by sponging miR-205-z in vitro, thereby playing a negative regulatory role in the antiviral immune response of rainbow trout. We also found that miR-205-z was a positive regulator of rainbow trout liver cell proliferation, and this effect could be reversed by SOCS3. In vivo, SOCS3 expression significantly increased after antagomiR-205-z injection. Furthermore, SOCS3 overexpression significantly promoted the replication of IHNV. This study provides fundamental data for disease resistance breeding and targeted drug therapy in rainbow trout.
Collapse
Affiliation(s)
- Lu Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jinqiang Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yongjuan Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- College of Science, Gansu Agricultural University, Lanzhou, China
| | - Shenji Wu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
5
|
Nicholls LA, Zeile KA, Scotto LD, Ryznar RJ. Timing of dietary effects on the epigenome and their potential protective effects against toxins. Epigenetics 2025; 20:2451495. [PMID: 39825851 DOI: 10.1080/15592294.2025.2451495] [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: 04/15/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/20/2025] Open
Abstract
Exposure to toxins causes lasting damaging effects on the body. Numerous studies in humans and animals suggest that diet has the potential to modify the epigenome and these modifications can be inherited transgenerationally, but few studies investigate how diet can protect against negative effects of toxins. Potential evidence in the primary literature supports that caloric restriction, high-fat diets, high protein-to-carbohydrate ratios, and dietary supplementation protect against environmental toxins and strengthen these effects on their offspring's epigenome. Most notably, the timing when dietary interventions are given - during a parent's early development, pregnancy, and/or lifetime - result in similar transgenerational epigenetic durations. This implies the existence of multiple opportunities to strategically fortify the epigenome. This narrative review explores how to best utilize dietary modifications to modify the epigenome to protect future generations against negative health effects of persistent environmental toxins. Furthermore, by suggesting an ideal diet with specific micronutrients, macronutrients, and food groups, epigenetics can play a key role in the field of preventive medicine. Based on these findings, longitudinal research should be conducted to determine if a high protein, high-fat, and low-carbohydrate diet during a mother's puberty or pregnancy can epigenetically protect against alcohol, tobacco smoke, and air pollution across multiple generations.
Collapse
Affiliation(s)
- Lynnea A Nicholls
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - Kendall A Zeile
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - London D Scotto
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - Rebecca J Ryznar
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
- Department of Biomedical Sciences, Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| |
Collapse
|
6
|
Gong Z, Zhang J, Yu J, Liu L, Wang X, Ma Q, Deng G, Wu X. Mycobacterium tuberculosis stimulates cuproptosis by regulating Lnc-Gm5532 to target FDX1 for bacteria intracellular survival. Int Immunopharmacol 2025; 160:114967. [PMID: 40449270 DOI: 10.1016/j.intimp.2025.114967] [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: 11/28/2024] [Revised: 04/08/2025] [Accepted: 05/25/2025] [Indexed: 06/03/2025]
Abstract
Cuproptosis is a novel cell death modality but its regulatory role in tuberculosis remains obscure. This study is committed to explore the specific mechanism of cuproptosis induced by Mycobacterium tuberculosis (M.tb) infection. We confirmed that M.tb induced cuproptosis contributes to its intracellular survival. Then microarray analysis was employed to screen M.tb-infected DE mRNAs and LncRNAs which may be potential regulators of cuproptosis. We focus on a key cuproptosis gene FDX1, and 7 LncRNAs that can target FDX1. Among them, LncRNA-Gm5532 upregulated the expression of FDX1 by sponging miR-7232-5p, which further promoted the expression of cuproptosis-associated proteins, increased intracellular copper ions, worsened lipid acylation-DLAT aggregation, and resulting in cuproptosis. Meanwhile, LncRNA-Gm5532 boosts the survival of M.tb in macrophages. Collectively, LncRNA-Gm5532 exacterbates M.tb-induced cuproptosis by targeting FDX1 and promotes immune escape of M.tb. The study will provide new insights into the regulation of tuberculosis pathogenesis.
Collapse
Affiliation(s)
- Zhaoqian Gong
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Jiaxue Zhang
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Jialin Yu
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Li Liu
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Xiaoping Wang
- The Fourth People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia 750021, China
| | - Qiumeng Ma
- Analvsis and Testing Center, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Guangcun Deng
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China.
| | - Xiaoling Wu
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China.
| |
Collapse
|
7
|
Fang Z, Pan Y, Lu Z, Wang L, Hu X, Ma Y, Li S. LncRNA SNHG1: A novel biomarker and therapeutic target in hepatocellular carcinoma. Gene 2025; 958:149462. [PMID: 40187618 DOI: 10.1016/j.gene.2025.149462] [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: 10/28/2024] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality globally. Increasing evidence suggests that long non-coding RNAs play a critical role in cancer development, with the small nucleolar RNA host gene family being a key participant in multiple types of carcinogenesis, including HCC. Small nucleolar RNA host gene 1 (SNHG1) is a significant member of the SNHG family. SNHG1 expression consistently increases in various HCC-associated processes, such as cell proliferation, apoptosis, angiogenesis, migration, invasion, and treatment resistance. Higher SNHG1 expression levels predict worse prognosis by positively correlating with clinicopathological features, including larger tumour size, poor differentiation, and advanced stages in patients with HCC. Nevertheless, the precise role of SNHG1 in the initiation and progression of HCC remains unclear. Therefore, this review aims to summarise the current investigations on the pathogenesis of SNHG1 in HCC, highlighting its potential as a molecular marker for early prediction and prognostic assessment. As a multifunctional modulator, SNHG1 is extensively involved in molecular signalling pathways in HCC progression and is valuable for therapeutic targeting.
Collapse
Affiliation(s)
- Zhou Fang
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, No.739 Dingshen Road, Zhoushan 316021 Zhejiang Province, China
| | - Yong Pan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou 31003, China
| | - Zhengmei Lu
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, No.739 Dingshen Road, Zhoushan 316021 Zhejiang Province, China
| | - Lingyun Wang
- Department of Infectious Diseases, Zhoushan Hospital, Zhejiang University, No.739 Dingshen Road, Zhoushan 316021 Zhejiang Province, China
| | - Xiaodan Hu
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, No.739 Dingshen Road, Zhoushan 316021 Zhejiang Province, China
| | - Yingqiu Ma
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, No.739 Dingshen Road, Zhoushan 316021 Zhejiang Province, China
| | - Shibo Li
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, No.739 Dingshen Road, Zhoushan 316021 Zhejiang Province, China.
| |
Collapse
|
8
|
Zhao L, Chang Y, Sun X, Chen H, Li N, Ma T, Jin S. LncRNA SNHG12 promotes EMT and metastasis of colorectal cancer via regulating TGF-β/Smad2/3 signaling pathway. Mol Immunol 2025; 183:225-235. [PMID: 40412277 DOI: 10.1016/j.molimm.2025.05.017] [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: 03/20/2025] [Revised: 04/21/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVE In this study, we aimed to explore the molecular mechanism of SNHG12 promoting colorectal cancer (CRC) progression. METHODS Bioinformatics technology was utilized to identify SNHG12-targeted mRNA and the correlation with the prognosis of CRC patients. Transfected sequence of knockdown SNHG12 in HCT-116 cell line was established. CCK8 assay, colone formation assay, flow cytometry, cell migration and transwell assay were applied to detect the impact of SNHG12 on HCT-116 cells. Besides, qRT-PCR and western blot were employed to evaluate the apoptotic and EMT markers as well as the expression of TGF-β and p-Smad2/3. Additionally, the rescue test of overexpressing TGF-β and a nude mouse subcutaneous tumor model were established to validate the pivotal role of SNHG12 in driving the progression of CRC. RESULTS SNHG12 could predict the prognosis of CRC patients, and a target mRNA GOLT1B was obtained from bioinformatics. In vitro results indicated that SNHG12 facilitated the proliferation, migration, and invasion of HCT-116 cells. qRT-PCR and western blot showed SNHG12 was related to the expression of Caspase 3, EMT markers as well as TGF-β and p-Smad2/3. Meanwhile, the rescue experiment proved that overexpressed TGF-β had the ability to reverse the impact of SNHG12 knockout on cell function and phenotype. In vivo, SNHG12 knockdown significantly reduced tumor growth. CONCLUSION SNHG12 promotes EMT and metastasis of CRC by modulating the TGF-β/Smad2/3 signaling pathway and EMT process, which could function as a prognostic biomarker and a treatment target for CRC.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Yuan Chang
- Department of Rheumatology and Immunology,The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Xiaoli Sun
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Hongliang Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Ning Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Tianyi Ma
- Department of Colorectal Surgery, Cancer Center,The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China.
| |
Collapse
|
9
|
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
|
10
|
Zhang H, Zhu Y, Yang C, Fu L, Huang X. LncRNA FOXD3-AS1 modulates ER stress and epithelial barrier dysfunction in allergic rhinitis by destabilizing CHOP mRNA. Cell Signal 2025; 131:111737. [PMID: 40081548 DOI: 10.1016/j.cellsig.2025.111737] [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: 01/13/2025] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Allergic rhinitis (AR) is an allergic disease of nasal mucosa. LncRNAs are key modulators affecting AR development. Nevertheless, the impact of LncRNA FOXD3-AS1 in AR is not clear. METHODS Human nasal epithelial cells (hNECs) were exposed to ovalbumin (OVA) to establish AR cell model, AR mice model was also constructed by OVA treatment. RIP assay was conducted to verify the association between FOXD3-AS1 and RBM15B. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses were performed to detect the expression of ER stress markers: Autophagy Related Gene 4 (ATG4), phosphorylated Protein kinase R-like endoplasmic reticulum kinase (p-PERK), and phosphorylated eukaryotic initiation factor 2α (p-eIF2α) in hNECs after overexpression of FOXD3-AS1. HNECs were treated with ER stress inhibitor 4-phenylbutyric acid (4-PBA). RESULTS The expressions of LncRNA FOXD3-AS1 were downregulated in AR model. Moreover, overexpression FOXD3-AS1 reversed the effect of AR on ER stress markers. RBM15B was found to be bound with FOXD3-AS1. After 4-PBA treatment, the protein expression of ATG4, CHOP, p-PERK, and p-eIF2α was significantly reduced in cultured AR cell model. CONCLUSION This study illustrated that FOXD3-AS1 acted as an inhibitor in AR induced ER stress and epithelial barrier dysfunction by destabilizing CHOP mRNA via RBM15B.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Otolaryngology Head and Neck surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Yaqiong Zhu
- Department of Otolaryngology Head and Neck surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chunping Yang
- Department of Otolaryngology Head and Neck surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Li Fu
- Department of Otolaryngology Head and Neck surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xinyi Huang
- Department of Otolaryngology Head and Neck surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
11
|
Ren S, Wang J, Wang Y, Luo Q, Pu W, Meng X, Liu S. Oxidative stress and type 2 diabetes: a review of lactic acid bacteria as potential prophylactic and therapeutic interventions. Food Sci Biotechnol 2025; 34:2403-2416. [PMID: 40492034 PMCID: PMC12145373 DOI: 10.1007/s10068-024-01775-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 06/11/2025] Open
Abstract
Oxidative stress, which results from the overproduction of reactive oxygen species (ROS) that induce protein, lipid, and DNA oxidation, has emerged as a key factor in the pathogenesis of various diseases, including type 2 diabetes (T2D). Recently, the relationship between oxidative stress and T2D has gained considerable attention. Widely utilized as probiotics in fermented foods and beverages, lactic acid bacteria (LAB) exhibit potent antioxidant properties. However, the precise mechanisms enabling LAB to behave as antioxidants remain elusive. LAB play a pivotal role in promoting and maintaining host health while mitigating the development and progression of various disorders, including T2D. Against the backdrop of a large number of studies highlighting the beneficial role of LAB in mitigating oxidative stress-related diseases, this review explores potential biomarkers for the prevention of oxidative stress and examines the potential contribution of LAB to the fight against T2D.
Collapse
Affiliation(s)
- Siheng Ren
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Juan Wang
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Yuxi Wang
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Qian Luo
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Wei Pu
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Xiao Meng
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Shukun Liu
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| |
Collapse
|
12
|
Zhang Y, Wang X, Luo H, Liu X, Huang J, Mi Z, He S, Wen J, Gao Q, Yang H, Feng Y, Li L, Zhai X, Wang F, Dai Y, Zhang L, Niu T, Sun C, Zheng Y. LncRNA HCG11 regulates selinexor sensitivity in multiple myeloma. Biochem Pharmacol 2025; 237:116948. [PMID: 40228640 DOI: 10.1016/j.bcp.2025.116948] [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: 01/18/2025] [Revised: 03/21/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Multiple myeloma (MM) is an incurable plasma cell caner featured by monoclonal plasma cell proliferation in bone marrow. The patients inevitably encounter drug resistance and the cancer relapse. Selinexor, a selective inhibitor of nuclear export by targeting exportin-1 (XPO1), is a novel medication in MM treatment. However, there were still many MM patients who did not respond to selinexor without mechanisms being fully demonstrated. In this study, we found that a long noncoding RNA (lncRNA), HLA complex Group 11 (HCG11), regulated MM cell growth and sensitivity to selinexor. MM cell lines and primary MM cell expressed HCG11. High HCG11 expression in MM correlated with inferior patients' overall survival. HCG11 knockdown (HCG11-KD) MM cells were more sensitive to selinexor-induced apoptosis. Mechanistically, HCG11 bound to XPO1 mRNA and promoted the mRNA stability and translation in cells. Thus, HCG11-KD MM cells had reduced XPO1 expression. Animal study showed that HCG11-KD MM was more sensitivity to selinexor than control knockdown in vivo. Overall, our study suggested a regulation of selinexor sensitivity in MM by lncRNA HCG11.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wang
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Luo
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Liu
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Jingcao Huang
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Ziyue Mi
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Siyao He
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Wen
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China; Department of Hematology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Qianwen Gao
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China; College of Life Science, Sichuan University, Chengdu, China
| | - Haonan Yang
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Feng
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Linfeng Li
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Zhai
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Fangfang Wang
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Dai
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Niu
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Yuhuan Zheng
- Department of Hematology/Institute of Hematology Research, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Wang Z, Su X, Zhan Z, Wang H, Zhou S, Mao J, Xu H, Duan S. miR-660: A novel regulator in human cancer pathogenesis and therapeutic implications. Gene 2025; 953:149434. [PMID: 40120868 DOI: 10.1016/j.gene.2025.149434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
MicroRNAs (miRNAs) are non-coding RNAs that regulate gene expression. Among these, miR-660, located on chromosome Xp11.23, is increasingly studied for its role in cancer due to its abnormal expression in various biological contexts. It is regulated by 8 competing endogenous RNAs (ceRNAs), which adds complexity to its function. miR- 660 targets 19 genes involved in 6 pathways such as PI3K/AKT/mTOR, STAT3, Wnt/β-catenin, p53, NF‑κB, and RAS, influencing cell cycle, proliferation, apoptosis, and invasion/migration. It also plays a role in resistance to chemotherapies like cisplatin, gemcitabine, and sorafenib in lung adenocarcinoma (LUAD), pancreatic ductal adenocarcinoma (PDAC), and hepatocellular carcinoma (HCC), thus highlighting its clinical importance. Additionally, leveraging liposomes as nanocarriers presents a promising avenue for enhancing cancer drug delivery. Our comprehensive study not only elucidates the aberrant expression patterns, biological functions, and regulatory networks of miR-660 and its ceRNAs but also delves into the intricate signaling pathways implicated. We envisage that our findings will furnish a robust framework and serve as a seminal reference for future investigations of miR-660, fostering advancements in cancer research and potentially catalyzing breakthroughs in cancer diagnosis and treatment paradigms.
Collapse
Affiliation(s)
- Zehua Wang
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xinming Su
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhiqing Zhan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hangxuan Wang
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shuhan Zhou
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Jiasheng Mao
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hening Xu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shiwei Duan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Tang D, Wang CF, Wang J, Jing XT, Ma J. Mechanism of the epidermal growth factor receptor in promoting endothelial cell dysfunction in gestational diabetes mellitus. World J Diabetes 2025; 16:105173. [DOI: 10.4239/wjd.v16.i6.105173] [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: 01/14/2025] [Revised: 03/29/2025] [Accepted: 05/08/2025] [Indexed: 06/13/2025] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) is a transmembrane protein that is differentially expressed in gestational diabetes mellitus (GDM). Endothelial dysfunction is a hallmark of GDM and plays a key role in its pathogenesis. EGFR is associated with endothelial dysfunction in the context of various diseases. However, the exact mechanism by which EGFR causes endothelial dysfunction in GDM is unknown, particularly its regulation at the transcriptional and protein levels.
AIM To explore the molecular mechanism by which EGFR influences endothelial cell dysfunction in GDM at the transcriptional and protein levels.
METHODS Quantitative real-time polymerase chain reaction was used to detect the expression of EGFR and H19. Western blotting was used to detect the expression of endothelial cell dysfunction markers. A cell counting kit 8 assay was used to assess cell viability, flow cytometry was used to assess apoptosis, scratch and Transwell assays were used to assess cell migration, and a tube formation assay was used to assess cell vascular formation. Hematoxylin-eosin staining was used to observe histopathological changes in the placentas of the mice.
RESULTS In this study, EGFR was upregulated in clinical samples, GDM animal models and GDM cell models, and the knockdown of EGFR could mitigate the effect of streptozotocin (STZ) and high glucose (HG); promoted the proliferation, migration and vascularization of human umbilical vein endothelial cells (HUVECs); inhibited cell apoptosis and the expression of endothelial cell dysfunction markers (vascular cell adhesion molecule-1, tumor necrosis factor-α, vascular endothelial growth factor-A, and intercellular cell adhesion molecule-1); and alleviated the process of GDM in vivo. Mechanistically, EIF4A3 binding to long noncoding RNA H19 increased the stability of EGFR messenger RNA, thereby promoting HG-induced HUVECs dysfunction or STZ-induced endothelial cell dysfunction in GDM mice. In addition, ERRFI1 also regulated the expression of EGFR, and ERRFI1 inhibited EGFR activity by binding to EGFR, thereby inhibiting HG-induced HUVECs dysfunction.
CONCLUSION Our study revealed that EGFR can accelerate the development of GDM by promoting endothelial cell dysfunction.
Collapse
Affiliation(s)
- Dan Tang
- Department of Obstetrics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Cheng-Fen Wang
- Department of Reproduction and Genetics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Jue Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Xiao-Tao Jing
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Jing Ma
- Department of Reproduction and Genetics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| |
Collapse
|
15
|
Chen Y, Dai H, Mao F, Li Y, Feng R, Qian Y. Long Noncoding RNA NONHSAT233728.1 Promotes ROS Accumulation and Granulosa Cell Apoptosis by Regulating the MAPK/ERK1/2 Signaling Pathway. FASEB J 2025; 39:e70681. [PMID: 40432414 PMCID: PMC12117357 DOI: 10.1096/fj.202500964r] [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: 03/26/2025] [Revised: 05/12/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most prevalent endocrine disorders in women of reproductive age. However, the underlying molecular mechanism remains unclear. In this study, we employed RNA sequencing analysis to identify differentially expressed protein-coding genes and long noncoding RNA (lncRNA) expression profiles in granulosa cells from women with and without PCOS. It was established that the level of NONHSAT233728.1 was diminished in women with PCOS. The present study demonstrated the role of NONHSAT233728.1 in granulosa cells from patients with PCOS and further investigated the potential mechanism of NONHSAT233728.1 in the KGN cell line. Additionally, the knockdown of NONHSAT233728.1 has been observed to promote cell apoptosis, inhibit cell proliferation, promote mitochondrial dysfunction, and inflammation. Western blot analyses confirmed that phospho-extracellular regulated protein kinases (ERK)1/2 were decreased following lnc-NONHSAT233728.1 knockdown. Consequently, we propose that ROS accumulation activates the endogenous mitochondrial apoptosis pathway, leading to granulosa cell apoptosis via the MEK/ERK1/2 pathway, which contributes to follicular atresia. We observed a negative correlation between NONHSAT233728.1 and both LH levels and the LH/FSH ratio. These findings indicate that lncRNA NONHSAT233728.1 is linked to the pathogenesis of PCOS and offer new insights into its underlying mechanisms.
Collapse
Affiliation(s)
- Yao Chen
- Reproductive Medical Center of Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Heqi Dai
- Reproductive Medical Center of Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Fei Mao
- Reproductive Medical Center of Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yangbai Li
- Reproductive Medical Center of Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Ruizhi Feng
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
- The Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yun Qian
- Reproductive Medical Center of Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
16
|
Olazagoitia-Garmendia A, Rojas-Márquez H, Trobisch T, Moreno-Castro C, Rodriguez Etxebarria A, Mentxaka J, Tripathi A, Yang B, Martin Ruiz I, Anguita J, Meana JJ, Ding Y, Dutta R, Schirmer L, Igoillo-Esteve M, Santin I, Castellanos-Rubio A. An inflammation-associated lncRNA induces neuronal damage via mitochondrial dysfunction. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102533. [PMID: 40291376 PMCID: PMC12023888 DOI: 10.1016/j.omtn.2025.102533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
Immune disease-associated non-coding SNPs, which often locate in tissue-specific regulatory elements, are emerging as key factors in gene regulation. Among these elements, long non-coding RNAs (lncRNAs) participate in many cellular processes, and their characteristics make these molecules appealing therapeutic targets. In this study, we have studied lncRNA LOC339803 in the context of neuronal cells, which is located in autoimmunity-associated region 2p15 and recently described to have a proinflammatory role in intestinal disorders. Using human brain samples and a wide variety of in vitro techniques, we have showed a differential function of this lncRNA in neuronal cells. We have further demonstrated the role of LOC339803 in maintaining hexokinase 2 (HK2) levels and thus mitochondrial integrity, partially explaining the implication of the lncRNA in multiple sclerosis (MS) pathogenesis. Our results show the importance of cell-type-specific studies in the case of regulatory lncRNAs. We present LOC339803 as a candidate for further studies as a mitochondrial dysfunction marker or possible therapeutic target in neurodegeneration.
Collapse
Affiliation(s)
- Ane Olazagoitia-Garmendia
- Department of Biochemistry and Molecular Biology, University of Basque Country UPV/EHU, 48940 Leioa, Spain
- Biobizkaia Health Research Institute, Cruces-Barakaldo 48903, Spain
| | - Henar Rojas-Márquez
- Biobizkaia Health Research Institute, Cruces-Barakaldo 48903, Spain
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Tim Trobisch
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Cristina Moreno-Castro
- ULB Center for Diabetes Research, Université Libre de Bruxelles (ULB), 1050 Bruxelles, Belgium
| | | | - Jon Mentxaka
- Department of Biochemistry and Molecular Biology, University of Basque Country UPV/EHU, 48940 Leioa, Spain
- Biobizkaia Health Research Institute, Cruces-Barakaldo 48903, Spain
| | - Ajai Tripathi
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland OH 44106, US
| | - Bibo Yang
- Department of Cell and Developmental Biology, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK
| | | | - Juan Anguita
- CIC bioGUNE-BRTA, 48160 Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - J Javier Meana
- Biobizkaia Health Research Institute, Cruces-Barakaldo 48903, Spain
- Department of Pharmacology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, 28029 Madrid, Spain
| | - Yiliang Ding
- Department of Cell and Developmental Biology, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK
| | - Ranjan Dutta
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland OH 44106, US
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69117 Heidelberg, Germany
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research, Université Libre de Bruxelles (ULB), 1050 Bruxelles, Belgium
| | - Izortze Santin
- Department of Biochemistry and Molecular Biology, University of Basque Country UPV/EHU, 48940 Leioa, Spain
- Biobizkaia Health Research Institute, Cruces-Barakaldo 48903, Spain
- CIBERDEM, 28029 Madrid, Spain
| | - Ainara Castellanos-Rubio
- Biobizkaia Health Research Institute, Cruces-Barakaldo 48903, Spain
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- CIBERDEM, 28029 Madrid, Spain
| |
Collapse
|
17
|
Zhang Z, Mao C, Wu Y, Wang Y, Cong H. Application of non‑coding RNAs in tumors (Review). Mol Med Rep 2025; 31:164. [PMID: 40211701 PMCID: PMC12015154 DOI: 10.3892/mmr.2025.13529] [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: 10/20/2024] [Accepted: 01/31/2025] [Indexed: 04/25/2025] Open
Abstract
Tumors are associated with the highest mortality rates worldwide. For more than a decade, research has focused on the genetic involvement of proteins in cancer; however, a complete class of molecular non‑coding (nc)RNAs have been discovered in recent years, and these are considered to be associated with cancer. Notably, ncRNAs are highly conserved and multifunctional. These interact with multiple signaling pathways, influencing cell cycle progression and various physiological processes. Therefore, the present review aimed to investigate ncRNA, microRNA, transfer RNA‑derived small RNA, PIWI‑interacting RNA and long non‑coding RNA to further understand the associated generation processes, functional mechanisms and therapeutic roles in tumors. The present review demonstrated the critical role of ncRNAs in tumors, and may provide a novel theoretical basis for the role of ncRNAs as biomarkers or therapeutic tools in the treatment of cancer.
Collapse
Affiliation(s)
- Zhihan Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Chunyan Mao
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yi Wu
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yin Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hui Cong
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Blood Transfusion, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
18
|
Cunha WR, Martin de la Vega M, Rodrigues de Barros P, Espinosa-Diez C. lncRNAs in vascular senescence and microvascular remodeling. Am J Physiol Heart Circ Physiol 2025; 328:H1238-H1252. [PMID: 40251747 DOI: 10.1152/ajpheart.00750.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/19/2024] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as critical regulators of vascular senescence and microvascular remodeling, processes that significantly contribute to the development of age-related diseases in organs such as the kidneys, heart, and lungs. Through mechanisms like chromatin remodeling, transcriptional regulation, and posttranscriptional modifications, lncRNAs modulate gene expression, thereby influencing cellular processes such as apoptosis, inflammation, fibrosis, and angiogenesis. In chronic kidney disease, cardiovascular disease, and pulmonary disorders, lncRNAs play a central role in promoting vascular dysfunction, endothelial cell aging, and fibrosis. This review focuses on how lncRNAs contribute to endothelial dysfunction, fibrosis, and vascular aging, emphasizing their roles in disease progression within the kidneys, heart, and lungs, where lncRNA-mediated vascular changes play a significant role in disease progression. Understanding the interactions between lncRNAs, vascular senescence, and microvascular remodeling offers promising avenues for developing targeted therapeutic strategies to mitigate the impact of aging on vascular health.
Collapse
Affiliation(s)
- Warlley Rosa Cunha
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Maria Martin de la Vega
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Paula Rodrigues de Barros
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Cristina Espinosa-Diez
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
19
|
Liu R, Xiao Y, Huang S, Wu H, Dong J, Zeng S, Li Y, Ye J, Wu W, Wang M, Zhang S, Lin Z, Song H. LncRNA XIST inhibits mitophagy and increases mitochondrial dysfunction by promoting BNIP3 promoter methylation to facilitate the progression of KBD. Mol Immunol 2025; 182:62-75. [PMID: 40179650 DOI: 10.1016/j.molimm.2025.03.016] [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: 12/24/2024] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
The primary mechanisms underlying cartilage destruction in Kashin-Beck disease (KBD) involve excessive chondrocyte death and extracellular matrix (ECM) degradation. While long non-coding RNA XIST (lncRNA XIST) has been implicated in promoting chondrocyte injury in osteoarthritis (OA), its role in KBD-related chondrocyte injury remains poorly understood. In this study, joint tissues were collected from four healthy and four KBD-affected children, as well as five healthy and five KBD-affected adults, to assess the expression of lncRNA XIST. The results revealed a significant upregulation of lncRNA XIST in the cartilage tissues of KBD patients. To model KBD-induced chondrocyte damage in vitro, hypertrophic ATDC5 cells were exposed to 10 ng/ml T-2 toxin for 24 hours, which resulted in increased lncRNA XIST expression. Silencing lncRNA XIST was found to mitigate T-2 toxin-induced ECM degradation and chondrocyte apoptosis by alleviating defects in mitochondrial autophagy and dysfunction. Mechanistically, lncRNA XIST promoted the methylation of the BNIP3 promoter by recruiting DNA methyltransferases (DNMTs) to the BNIP3 promoter region, thereby suppressing BNIP3-mediated mitophagy and exacerbating mitochondrial dysfunction. To establish a KBD rat model, rats were fed a low-selenium diet supplemented with T-2 toxin for four weeks. Knockdown of lncRNA XIST in these rats attenuated articular cartilage damage and apoptosis, while enhancing collagen II expression. In conclusion, lncRNA XIST accelerates KBD progression by inhibiting mitophagy and promoting mitochondrial dysfunction through increased BNIP3 promoter methylation.
Collapse
Affiliation(s)
- Ruoxi Liu
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Xiao
- Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, China
| | - Sihua Huang
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Wu
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jun Dong
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Sixiang Zeng
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongwei Li
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jintao Ye
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wei Wu
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengxin Wang
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Sanpeng Zhang
- Operating room, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhaoxing Lin
- Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, China.
| | - Huanjin Song
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
20
|
Dong L, Wu H, Qi F, Xu Y, Chen W, Wang Y, Cai P. Non-coding RNA-mediated granulosa cell dysfunction during ovarian aging: From mechanisms to potential interventions. Noncoding RNA Res 2025; 12:102-115. [PMID: 40144342 PMCID: PMC11938093 DOI: 10.1016/j.ncrna.2025.03.001] [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: 09/01/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
As the earliest aging organ in the reproductive system, the ovary has both reproductive and endocrine functions, which are closely related to overall female health. The exact pathogenesis of ovarian aging (OA) remains incompletely understood, with granulosa cells (GCs) dysfunction playing a significant role in this process. Recent advancements in research and biotechnology have highlighted the importance of non-coding RNAs (ncRNAs), including micro RNAs, long non-coding RNAs, and circular RNAs, in regulating the biological functions of GCs through gene expression modulation. This paper provides a comprehensive overview of the role of ncRNAs in various cellular functions such as apoptosis, autophagy, proliferation, and steroid synthesis in GCs, and explores the underlying regulatory mechanisms. Additionally, the therapeutic potential of ncRNAs, particularly those carried by exosomes derived from mesenchymal stem cells, in delaying OA is discussed. Understanding the regulatory mechanisms of ncRNAs in GC function and the current progress in this field is crucial for identifying effective biomarkers and therapeutic targets, ultimately aiding in the early diagnosis, prognostic assessment, and individualized treatment of OA.
Collapse
Affiliation(s)
- Li Dong
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haicui Wu
- Department of Reproduction and Genetics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fanghua Qi
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuan Xu
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wen Chen
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuqi Wang
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Pingping Cai
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
21
|
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
|
22
|
Wang X, Wang Q, Wang H, Cai G, An Y, Liu P, Zhou H, Chen HW, Ji S, Ye J, Wang J. Small protein ERSP encoded by LINC02870 promotes triple negative breast cancer progression via IRE1α/XBP1s activation. Cell Death Differ 2025; 32:1014-1025. [PMID: 39799200 PMCID: PMC12162857 DOI: 10.1038/s41418-025-01443-5] [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: 01/25/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
Clinical treatment options for triple-negative breast cancer (TNBC) are currently limited to chemotherapy because of a lack of effective therapeutic targets. Recent evidence suggests that long noncoding RNAs (lncRNAs) encode bioactive peptides or proteins, thereby playing noncanonical yet significant roles in regulating cellular processes. However, the potential of lncRNA-translated products in cancer progression remains largely unknown. In this study, we identified a previously undocumented small protein encoded by the lncRNA LINC02870. This protein is localized at the endoplasmic reticulum (ER) and participates in ER stress, thus, we named it the endoplasmic reticulum stress protein (ERSP). ERSP was highly expressed in TNBC tissues, and elevated LINC02870 content was correlated with poor prognosis in TNBC patients. Loss of ERSP inhibited TNBC growth and metastasis both in vitro and in vivo. The pro-oncogenic effects of ERSP could be attributed to its selective activation of the IRE1α/XBP1s branch. ERSP enhances the unfolded protein response (UPR) by interacting with XBP1s, facilitating the nuclear accumulation of XBP1s, thereby promoting the expression of ER stress-related genes. These findings highlight the regulatory role of the lncRNA-encoded protein ERSP in ER stress and suggest that it is a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Xiaolu Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Qianqian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Hong Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Guodi Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Yana An
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Peiqing Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Huihao Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Shufeng Ji
- Special Medical Service Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510006, China.
| | - Jiantao Ye
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
| | - Junjian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
23
|
Li Z, Liu X, Tang X, Yang Y. Analysis of gonadal transcriptome reveals core long non-coding RNA-mRNA regulatory network in sea cucumber Apostichopus japonicus. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101396. [PMID: 39667089 DOI: 10.1016/j.cbd.2024.101396] [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: 09/04/2024] [Revised: 11/26/2024] [Accepted: 12/08/2024] [Indexed: 12/14/2024]
Abstract
Apostichopus japonicus is a representative temperate sea cucumber species, that mainly inhabits in coastal zone of the continental shelf. With high nutritional value and important medical value, A. japonicus become an important commercial aquaculture species and produce significant economic value in recent years. A. japonicus has no sexual dimorphism that can be used to distinguish female and male individuals by external appearance and morphology. The phenotype sex can be only detected by dissecting and observing gonad tissue, thus the breeding efficiency could be greatly reduced. This limitation has hindered the advancement of selective breeding programs and sea cucumber industry. To investigate the genetic basis of reproductive biology in A. japonicus, advanced sequencing techniques, such as next- and third-generation sequencing, have been employed to explore the roles of non-coding RNAs and other genetic factors, offering new insights into sex determination mechanisms. To further gain a deeper understanding of the knowledge underlying lncRNAs in gonadal differentiation, we conducted a comparative transcriptome sequencing analysis of gonadal tissues from both sexes. In our research, a total of 3990 novel lncRNAs and 1441 differentially expressed lncRNAs were identified between female and male gonads. Additionally, a molecular regulatory network indicating lncRNA-mRNA interactions was constructed based on transcriptional profiles, which provide insights into the potential cis- and trans- target genes of lncRNAs. The gonadal transcriptome analysis identified a number of novel long non-coding RNAs involved in female and male reproduction process. Both cis- and trans-acting regulatory networks indicating lncRNA-mRNA interaction were constructed based on transcriptional profiles. These findings provide new insights into the lncRNA-mediated regulation of reproductive biology in marine invertebrates, indicating the crucial roles of long non-coding sequences in regulating expression profiles. Further, the GO and KEGG enrichment analyses of cis- and trans- targeted mRNA for differentially expressed lncRNA indicated that sexual reproduction (GO:0019953), germ cell development (GO:0007281), and negative regulation of hormone secretion (GO:0046888) are potentially involved in gonadal differentiation through the regulation of long non-coding sequences. Notably, besides the classical reproduction related signaling pathway like Gonadotropin-releasing hormone (GnRH) secretion (ko04929), several regulatory pathways, such as Epidermal growth factor receptor (ErbB) signaling pathway (ko04012), TGF-beta signaling pathway (ko04350), and neurotrophin signaling pathway (ko04722) were also enriched and potentially involved in sex differentiation and gonadal development.
Collapse
Affiliation(s)
- Ziming Li
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Xinghai Liu
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Xinyue Tang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Yujia Yang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China.
| |
Collapse
|
24
|
Zhu X, Mao Z, Zheng P, Wang L, Zhang F, Zi G, Liu H, Zhang H, Liu W, Zhou L. The role and research progress of epigenetic modifications in obstructive sleep apnoea-hypopnea syndrome and related complications. Respir Med 2025; 242:108099. [PMID: 40228610 DOI: 10.1016/j.rmed.2025.108099] [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: 12/05/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Epigenetic modifications are heritable changes in gene expression that do not alter the DNA sequence. Histone modifications, non-coding RNA expression, and DNA methylation are examples of common epigenetic changes. Obstructive sleep apnoea-hypopnea syndrome (OSAHS) is the most common sleep-related breathing disorder, and its incidence is increasing annually, making it a hotspot of clinical research and significantly impacting health and well-being. The main cause of OSAHS is related to complications caused by repeated chronic intermittent hypoxia (CIH). Currently, polysomnography (PSG) and continuous positive airway pressure (CPAP) remain the gold standards for the diagnosis and treatment of OSAHS. However, their limitations-such as time consumption, high cost, and poor patient comfort-contribute to the paradox of high disease prevalence yet low rates of diagnosis and treatment, resulting in a substantial disease burden. In recent years, rapid advances in epigenetics have revealed that biomarkers such as microRNAs (miRNAs), circular RNAs (circRNAs), and other epigenetic modifications hold promise as non-invasive tools for the diagnosis and treatment of OSAHS and its related complications. Although numerous studies have explored epigenetic modifications in other diseases, this study focuses on how epigenetic modifications participate in the process of OSAHS and its related complications, with an aim of elucidating the pathogenesis of OSAHS from an epigenetic perspective and provide new directions for identifying molecular targets for the diagnosis and treatment of OSAHS and related complications.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhenyu Mao
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pengdou Zheng
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lingling Wang
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengqin Zhang
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guisha Zi
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huojun Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Wei Liu
- Department of Geriatrics, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China.
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
25
|
Cheng L, Yu H, Qin Y, Wang R, Su T, Lyu G, Huang Z, Li H, Jin Y, Li Y, Wang G. The lncRNA ST18-AS1 suppresses pancreatic cancer progression by enhancing ST18 mRNA stability through anchoring FUS in the cytoplasm. Oncogene 2025:10.1038/s41388-025-03455-4. [PMID: 40450113 DOI: 10.1038/s41388-025-03455-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 05/10/2025] [Accepted: 05/19/2025] [Indexed: 06/03/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with a high mortality rate and short survival time. Long noncoding RNAs (lncRNAs) play a significant role in the progression of PDAC. However, prognostic lncRNAs associated with overall survival (OS) in patients with PDAC remain elusive. RNA sequencing was used to identify differential lncRNA expression between short-term and long-term PDAC patients. We identified a novel lncRNA (ENSG00000253924), termed ST18-AS1 (ST18-associated lncRNA), that is highly expressed in the tissues of long-term PDAC patients. High ST18-AS1 expression was correlated with favorable clinical outcomes, and the upregulation of ST18-AS1 expression in PDAC cell lines suppressed cell proliferation and promoted apoptosis both in vivo and in vitro. The key downstream target regulated by ST18-AS1 was Suppression of tumorigenicity 18 (ST18). We also found that ST18 expression was lower in PDAC tissues compared to non-tumorous adjacent tissues (NATs) and that higher ST18 expression was correlated with better clinical outcomes. Accordingly, the forced expression of ST18 inhibited proliferation and promoted apoptosis in tumor cells. Mechanistic studies showed that ST18-AS1 maintained the stability of ST18 mRNA by binding to Fused in sarcoma (FUS) and anchoring FUS in the cytoplasm. Overall, we identified ST18-AS1 as a novel biomarker that inhibits PDAC cell proliferation and promotes PDAC cell apoptosis through ST18. Targeting ST18-AS1/ST18 may be a potential therapeutic strategy for treating PDAC.
Collapse
Affiliation(s)
- Long Cheng
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hang Yu
- Department of General Surgery, Harbin The First Hospital, Harbin, China
| | - Yujing Qin
- Department of Gastroenterology, Heilongjiang Provincial Hospital, Harbin, China
- Department of Gastroenterology, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin, China
| | - Ruixin Wang
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tie Su
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gaoyuan Lyu
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zijian Huang
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongtao Li
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Jin
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Gang Wang
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
26
|
Kadeerbieke B, Wu L, Zhang YM. The role of lncRNA H19/Hmox1 axis regulating ferroptosis in anthracycline-induced cardiotoxicity. Drug Chem Toxicol 2025:1-12. [PMID: 40432412 DOI: 10.1080/01480545.2025.2503946] [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: 11/11/2024] [Revised: 04/24/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025]
Abstract
This study investigates the molecular mechanisms underlying anthracyclines (ANT)-induced cardiotoxicity, with a specific focus on ferroptosis regulated by the long non-coding RNA (lncRNA) H19/heme oxygenase-1 (Hmox1) signaling axis. A retrospective analysis was performed on 50 breast cancer patients who developed ANT-associated cardiac dysfunction. Clinical assessments included measurements of left ventricular ejection fraction (LVEF) and serum markers, such as cardiac troponin I (cTnI), creatine kinase-MB (CK-MB), N-terminal pro-B-type natriuretic peptide (NT-proBNP), and serum iron levels. Serum analysis revealed a marked downregulation of lncRNA H19 and upregulation of Hmox1, both significantly correlated with impaired cardiac function and disrupted iron homeostasis. To further elucidate the mechanism, an Epirubicin (EPI)-induced injury model in HL-1 cardiomyocytes was established. EPI exposure led to suppression of lncRNA H19, upregulation of Hmox1, and induction of apoptosis and ferroptotic cell death. RNA-seq analysis identified potential downstream targets linking lncRNA H19 to iron metabolism via Hmox1 modulation. Functional assays demonstrated that overexpression of lncRNA H19 mitigated EPI-induced ferroptosis, while enforced expression of Hmox1 reversed these protective effects. Collectively, these findings identify the lncRNA H19/Hmox1 axis as a critical regulator of ferroptosis in ANT-induced cardiotoxicity and suggest it as a potential therapeutic target for mitigating cardiac injury in breast cancer patients undergoing anthracycline chemotherapy.
Collapse
Affiliation(s)
- Bayan Kadeerbieke
- Department of Oncology Cardiology, Xinjiang Medical University Cancer Hospital, Urumqi, China
| | - Li Wu
- Department of Oncology Cardiology, Xinjiang Medical University Cancer Hospital, Urumqi, China
| | - Yuan-Ming Zhang
- Department of Oncology Cardiology, Xinjiang Medical University Cancer Hospital, Urumqi, China
| |
Collapse
|
27
|
Jing Q, Chen Q, Wang G, Wu T, Wang L, Xiong Q, Yang X, Qiu L, Han J. LINC02593 impedes cell senescence via COP1-mediated p53 degradation in cervical cancer. Cell Signal 2025; 134:111907. [PMID: 40441468 DOI: 10.1016/j.cellsig.2025.111907] [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: 03/28/2025] [Revised: 05/15/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025]
Abstract
Evasion of cellular senescence is one of the hallmarks of cervical carcinoma (CC) to maintain malignant development. Even though the regulators driving CC cell senescence are widely recognized, the underlying upstream mechanisms are still not fully understood. Long non-coding RNAs (lncRNAs) are emerging as important regulators in cell senescence. Here, we conducted a lncRNA profiling and identified LINC02593 as a significantly downregulated lncRNA in induced senescent cervical squamous cell carcinoma (CSCC) cells. LINC02593 is upregulated in CSCC tissues. Depletion of LINC02593 resulted in a marked cellular senescence phenotype and tumor growth inhibition in vitro and in vivo, whereas LINC02593 overexpression suppressed doxorubicin-induced cell senescence. LINC02593 was shown to impede cell senescence by inhibiting p21 expression, and this regulation was mainly dependent on p53 protein degradation. Mechanistically, LINC02593 served as a scaffold, bridging the coiled-coil domain of COP1 and the C-terminal domain of p53, enhancing the affinity between p53 and its E3 ubiquitin ligase COP1. The "scaffold" function facilitated p53 degradation by COP1 as well as the downstream p21 repression, eventually evading cell senescence. Overall, we characterized a previously unknown mechanism by which LINC02593 manipulated senescence to promote CC progression.
Collapse
Affiliation(s)
- Qian Jing
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qin Chen
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guosong Wang
- Laboratory of Gastrointestinal Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tong Wu
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lingli Wang
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qunli Xiong
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaojuan Yang
- Abdominal Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Qiu
- Laboratory of Gastrointestinal Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610093, China.
| |
Collapse
|
28
|
Choudhury S, Mehta NK, Raghava GPS. CytoLNCpred-a computational method for predicting cytoplasm associated long non-coding RNAs in 15 cell-lines. FRONTIERS IN BIOINFORMATICS 2025; 5:1585794. [PMID: 40491847 PMCID: PMC12146324 DOI: 10.3389/fbinf.2025.1585794] [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: 03/01/2025] [Accepted: 05/14/2025] [Indexed: 06/11/2025] Open
Abstract
The function of long non-coding RNA (lncRNA) is largely determined by its specific location within a cell. Previous methods have used noisy datasets, including mRNA transcripts in tools intended for lncRNAs, and excluded lncRNAs lacking significant differential localization between the cytoplasm and nucleus. In order to overcome these shortcomings, a method has been developed for predicting cytoplasm-associated lncRNAs in 15 human cell-lines, identifying which lncRNAs are more abundant in the cytoplasm compared to the nucleus. All models in this study were trained using five-fold cross validation and tested on an validation dataset. Initially, we developed machine and deep learning based models using traditional features like composition and correlation. Using composition and correlation based features, machine learning algorithms achieved an average AUC of 0.7049 and 0.7089, respectively for 15 cell-lines. Secondly, we developed machine based models developed using embedding features obtained from the large language model DNABERT-2. The average AUC for all the cell-lines achieved by this approach was 0.665. Subsequently, we also fine-tuned DNABERT-2 on our training dataset and evaluated the fine-tuned DNABERT-2 model on the validation dataset. The fine-tuned DNABERT-2 model achieved an average AUC of 0.6336. Correlation-based features combined with ML algorithms outperform LLM-based models, in the case of predicting differential lncRNA localization. These cell-line specific models as well as web-based service are available to the public from our web server (https://webs.iiitd.edu.in/raghava/cytolncpred/).
Collapse
|
29
|
Zhang R, Zhang Y, Li X, Wang S, Li J, Shao S, Liu B, Guo W, Shi M. Long non-coding RNA HOXA11-AS inhibits apoptosis, induces proliferation, and promotes autophagy via the miR-214-3p/ATG12 axis in acute T lymphoblastic leukemia. J Mol Histol 2025; 56:170. [PMID: 40418508 DOI: 10.1007/s10735-025-10462-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 05/09/2025] [Indexed: 05/27/2025]
Abstract
Acute T lymphocytic leukemia (T-ALL) is a hematological cancer with high mortality. The literature suggests an association between T-ALL and Long non-coding RNAs (lncRNAs). Nevertheless, the mechanism of lncRNA HOXA11-AS in T-ALL remains undetermined. The lncRNA HOXA11-AS expression level were assessed in T-ALL patients and normal controls by qRT-PCR. Furthermore, The proliferation, apoptosis, and autophagy of T-ALL cell (Molt4 and Jurkat cells) were investigated in vitro via EdU incorporation experiment, flow cytometry, immunofluorescence, and Western blotting. Moreover, the relationship between HOXA11-AS and miR-214-3p and between miR-214-3p, and ATG12 was verified via dual-luciferase reporter assays. Our investigation demonstrated that compared to normal controls, the HOXA11-AS expression level were increased in T-ALL patients. Furthermore, in T-ALL cells, inhibition of HOXA11-AS or overexpression of miR-214-3p reduced autophagy and proliferation, while stimulating apoptosis. However, miR-214-3p inhibition counteracted HOXA11-AS-mediated suppression of T-ALL cell proliferation, autophagy, and apoptosis. In addition, HOXA11-AS modulated ATG12 via sponging miR-214-3p. Overall, this research indicated that lncRNA HOXA11-AS not only stimulates cell proliferation and autophagy but also inhibits apoptosis via the miR-214-3p/ATG12 axis, thereby suggesting that lncRNA HOXA11-AS might be a new candidate for T-ALL diagnosis and therapy.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - YuanYuan Zhang
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - XiaoFei Li
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - ShaoHua Wang
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - Jiao Li
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - ShuoChun Shao
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - Bin Liu
- Department of Clinical Laboratory, Children's Hospital of Hebei Province, Shijiazhuang, 050031, People's Republic of China
| | - WeiWei Guo
- Department of Second Clinical Laboratory Children's Hospital of Hebei Province, Shijiazhuang, 050031, People's Republic of China
| | - Min Shi
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China.
- Hebei Key Laboratory of Laboratory Medicine, Shijiazhuang, 050017, People's Republic of China.
| |
Collapse
|
30
|
Chellini L, Del Verme A, Riccioni V, Paronetto MP. YAP1 promoter-associated noncoding RNA affects Ewing sarcoma cell tumorigenicity by regulating YAP1 expression. Cell Mol Biol Lett 2025; 30:63. [PMID: 40414844 PMCID: PMC12103783 DOI: 10.1186/s11658-025-00736-4] [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: 11/14/2024] [Accepted: 04/22/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Ewing sarcomas (ESs) are aggressive paediatric tumours of bone and soft tissues afflicting children and adolescents. Despite current therapies having improved the 5-year survival rate to 70% in patients with localized disease, 25% of patients relapse and most have metastasis at diagnosis. Resistance to chemotherapy, together with the high propensity to metastasize, remain the main causes of treatment failure. Thus, identifying novel targets for alternative therapeutic approaches is urgently needed. METHODS Biochemical and functional analyses were carried out to elucidate the mechanism of regulation of YAP1 expression by pncRNA_YAP1-1 in ES cells. RESULTS Here, we identified a novel promoter-associated noncoding RNA, pncRNA_YAP1-1, transcribed from the YAP1 promoter in ES cells. We found that pncRNA_YAP1-1 level exerts antitumour effects on ES by destabilizing YAP1 protein. The molecular mechanism relies on the interaction of pncRNA_YAP1-1 with the RNA binding protein FUS, which stabilizes the transcript. Furthermore, pncRNA_YAP1-1 binding to TEAD impairs its interaction with YAP1, thus determining YAP1 translocation into the cytoplasm, its phosphorylation and degradation. CONCLUSIONS Overall, our findings reveal a novel layer of regulation of YAP1 protein expression by pncRNA_YAP1-1 in Ewing sarcoma. Considering the role of YAP1 in therapy response and cell propensity to metastasize, our results indicate pncRNA_YAP1-1 as an actionable target that could be exploited to enhance chemotherapy efficacy in Ewing sarcoma. SIGNIFICANCE PncRNA_YAP1-1 counteracts the YAP1 oncogenic transcriptional program in Ewing sarcoma cells by interfering with YAP1-TEAD interaction and impairing YAP1 protein stability. These findings uncover a novel treatment option for Ewing sarcoma.
Collapse
MESH Headings
- Sarcoma, Ewing/genetics
- Sarcoma, Ewing/pathology
- Sarcoma, Ewing/metabolism
- Humans
- YAP-Signaling Proteins
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Promoter Regions, Genetic/genetics
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Bone Neoplasms/genetics
- Bone Neoplasms/pathology
- RNA-Binding Protein FUS/metabolism
- Carcinogenesis/genetics
- Carcinogenesis/pathology
Collapse
Affiliation(s)
- Lidia Chellini
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Santa Lucia Foundation, Rome, Italy.
| | - Arianna Del Verme
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Veronica Riccioni
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Paola Paronetto
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| |
Collapse
|
31
|
Fan D, Shang Y, Cong Y, Jiao Y, Li N, Zhao H. Reciprocal regulation between m6 A modifications and non-coding RNAs: emerging roles in cancer therapeutic resistance. Discov Oncol 2025; 16:920. [PMID: 40413672 DOI: 10.1007/s12672-025-02641-w] [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: 03/05/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
In recent years, the interplay between N6-methyladenosine (m6A) modifications and non-coding RNAs (ncRNAs) has emerged as a pivotal research area, owing to their crucial involvement in the pathophysiological mechanisms underlying various diseases. A significant hurdle in cancer therapy is therapeutic resistance, which frequently contributes to adverse patient outcomes. Recent investigations have underscored the vital role that interactions between m6A modifications and ncRNAs play in mediating cancer therapeutic resistance via the MAPK, PI3K/Akt/mTOR, Wnt/β-catenin, HIPPO, and NF-κB pathways. This review elucidates how these interactions drive tumor therapeutic resistance by modulating these pathways. By dissecting the regulatory dynamics between m6A and ncRNAs in the context of cancer therapeutic resistance, this review aims to deepen the understanding of m6A-ncRNA interaction in cancer therapeutic resistance and identify potential therapeutic targets to improve cancer treatment efficacy.
Collapse
Affiliation(s)
- Dan Fan
- Nanshan Class, The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Yan Shang
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Yating Cong
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Yanlin Jiao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Na Li
- The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Hailong Zhao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
32
|
Hu S, Chen G, Luo A, Zhao H, Li D, Peng B, Du J, Luo D. Mechanism of LINC01018/miR-182-5p/Rab27B in the immune escape through PD-L1-mediated CD8 + T cell suppression in glioma. Biol Direct 2025; 20:61. [PMID: 40399992 PMCID: PMC12093642 DOI: 10.1186/s13062-025-00651-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: 12/25/2024] [Accepted: 04/15/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Glioma is a malignant tumor associated with poorer prognosis. This study aims to elucidate the mechanism of LINC01018/miR-182-5p/Rab27B axis in PD-L1-mediated CD8+ T cell suppression in the progression of gliomas. METHODS LINC01018, miR-182-5p, and Rab27B expression levels in glioblastoma tissues were measured. The proportion of infiltrating macrophages and monocytes and CD8+ T cell function were assessed. The relationship between miR-182-5p and Rab27B was analyzed. Glioma cell activity, invasion, and migration were measured. The expression of E-cadherin, N-cadherin, Vimentin, PD-L1, iNOS, and CD206 was determined. Glioma cell-derived EVs were isolated, and the co-localization of Rab27B and PD-L1 and the binding of Rab27B to PD-L1 were analyzed. The endocytosis of EVs by microglia was assayed. The impact of LINC01018/miR-182-5p/Rab27B on glioma growth was observed. The function of macrophages and CD8+ T cells in tumors was analyzed. RESULTS Rab27B was downregulated, and infiltrating macrophages and monocytes were increased in glioblastoma. miR-182-5p inhibited Rab27B expression. Rab27B knockdown reverses the inhibitory effect of LINC01018 overexpression on glioma cell growth. Glioma cells-derived EVs with low Rab27B expression carried more PD-L1 to increase PD-L1 expression and M2 polarization in microglia. LINC01018 overexpression reduced macrophages in orthotopic tumors. CD8+ T cell numbers showed no significant difference, but TIM-3 increased and IFN-γ decreased. miR-182-5p inhibition enhanced the therapeutic effect of anti-PD-L1, which was reversed after glioma cell-derived EVs. CONCLUSION LINC01018 promotes PD-L1-mediated CD8+ T cell suppression via the miR-182-5p/Rab27B axis in glioma cell-derived EVs, thereby contributing to immune escape in gliomas.
Collapse
Affiliation(s)
- Su Hu
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Guoshuo Chen
- Department of Intervention, the Affiliated Cancer Hospital, Guangzhou Institute of Cancer Research, Guangzhou Medical University, Guangzhou, 510095, China
| | - Aiping Luo
- Department of Radiology, the Affiliated Cancer Hospital, Guangzhou Institute of Cancer Research, Guangzhou Medical University, Guangzhou, 510095, China
| | - Hailin Zhao
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Dan Li
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Biao Peng
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| | - Jike Du
- Department of Medical Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| | - Dongdong Luo
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| |
Collapse
|
33
|
Song X, Yao L, Li Y, Wang J, Lu C, Li J, Leng Q, Tang X, Hu X, Wu J, Chen R, Lin X, Ye J, Kuang X, Zhang G, Sun M, Zhou Y, Li H. Lnc-DARVR/miR-365-1-5p/LAMB1 axis regulates rotavirus replication via the complement C3 pathway. J Virol 2025; 99:e0211424. [PMID: 40237496 PMCID: PMC12090790 DOI: 10.1128/jvi.02114-24] [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: 01/22/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Antiviral effectors and cytokines are critical components of host innate immunity. However, the regulatory mechanisms governing the roles of these molecules in host-virus interactions are still unclear. Although long non-coding RNAs (lncRNAs) have been recognized as key players in various biological processes, their involvement in the complement system of host antiviral defenses remains to be explored. In this study, we discovered a novel, unannotated lncRNA, called DARVR. DARVR was found to be an intergenic lncRNA and inhibited rotavirus (RV) replication in MA104 cells. Mechanistically, we found that complement 3 (C3) was upregulated following RV infection in a LAMB1-dependent manner. However, LAMB1 expression was downregulated by miR-365-1-5p, resulting in the inhibition of the C3-mediated antiviral reaction. However, DARVR functioned as a competing endogenous RNA against miR-365-1-5p, promoting the expression of LAMB1 and thereby enhancing C3 activity and inhibiting RV replication. These results not only provide evidence demonstrating the involvement of lncRNAs in the regulation of RV infection but also highlight the role of complement factors in host innate immunity. IMPORTANCE Long non-coding RNAs (lncRNAs) play versatile and critical roles in host-virus interactions, offering significant potential for developing targeted therapies to prevent or treat viral infections. Despite their importance, the involvement of lncRNAs in rotavirus infection remains underexplored. This study identifies a novel lncRNA that enhances complement factor C3 activity through the competing endogenous RNA (ceRNA) mechanism, effectively inhibiting rotavirus replication across different subtypes. These findings underscore the complex molecular interplay regulating complement factor activity during rotavirus infection and provide valuable insights into the host's antiviral mechanisms. This research paves the way for innovative therapeutic strategies targeting lncRNAs and complement factors to combat viral infections more effectively.
Collapse
Affiliation(s)
- Xiaopeng Song
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Lida Yao
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Yan Li
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Jinlan Wang
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Chenxing Lu
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Jinmei Li
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Qingmei Leng
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Xianqiong Tang
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Xiaoqing Hu
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Jinyuan Wu
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Rong Chen
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Xiaochen Lin
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Jun Ye
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Xiangjun Kuang
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Guangming Zhang
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Maosheng Sun
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Yan Zhou
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| | - Hongjun Li
- Institute of Medical Biology, Peking Union Medical College Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Chinese Academy of Medical Sciences, Kunming, China
| |
Collapse
|
34
|
Gao M, Yin L, Zhang B, Dong Z, Jiang W, Bai Z, Zhao X, Xu L, Wang N, Peng J. Targeting Ischemic Myocardium: Nanoparticles Loaded with Long Noncoding RNA AK156373 siRNA Alleviate Myocardial Infarction. ACS NANO 2025; 19:18475-18491. [PMID: 40338223 DOI: 10.1021/acsnano.5c01641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Despite advancements in the development of targeted approaches for the treatment of myocardial infarction (MI), there is a continuing need for improvements in treatment approaches due to the high mortality and prevalence of MI. The identification of specific therapeutic targets and the development of efficient delivery systems are essential. In this study, a nanoparticle delivery system targeting necrotic cardiomyocytes was engineered. This system effectively downregulated long noncoding RNA (lncRNA) AK156373 and reduced oxidative stress and inflammation during MI progression. Mechanistically, silencing lncRNA AK156373 enhanced the viability and mitochondrial function of hypoxic cardiomyocytes and lowered intracellular inflammatory cytokine levels and reactive oxygen species (ROS) production. In vivo, cardiac-specific lncRNA AK15673 knockout mice were generated (AK156373flox/flox, Myh6-Cre mice), and lncRNA AK156373 knockout obviously reduced the infarct size, collagen fiber deposition, and ischemia severity in MI mice, leading to improved cardiac function. Additionally, lncRNA AK156373 modulated miR-204-5p to regulate C-X-C motif chemokine receptor 2 (CXCR2) protein expression via the competing endogenous RNA (ceRNA) mechanism, exacerbating myocardial damage and accelerating MI progression. Subsequently, nanoparticles loaded with lncRNA AK156373 siRNA were synthesized. The nanoparticles significantly inhibited MI progression by modulating the miR-204-5p/CXCR2 axis to reduce oxidative stress and inflammation. Overall, these findings establish a key regulatory role for lncRNA AK156373 in MI progression and present a direct preclinical approach for MI therapy.
Collapse
Affiliation(s)
- Meng Gao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Bo Zhang
- Department of Cardiology, The First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Zhichao Dong
- Department of Cardiology, The First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Wenjiao Jiang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Zhuoya Bai
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Xuerong Zhao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ning Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430065, China
| |
Collapse
|
35
|
Wang YJ, Zhi ZZ, Liu T, Kang J, Xu GH. Long non-coding RNA GAS5 promotes neuronal apoptosis in spinal cord injury via the miR-21/PTEN axis. World J Orthop 2025; 16:106183. [DOI: 10.5312/wjo.v16.i5.106183] [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: 02/18/2025] [Revised: 03/18/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a severe and permanent trauma that often leads to significant motor, sensory, and autonomic dysfunction. Neuronal apoptosis is a major pathomechanism underlying secondary injury in SCI. Long non-coding RNAs (lncRNAs) have emerged as key regulators of gene expression and cellular processes, including apoptosis. However, the role of lncRNA growth arrest-specific transcript 5 (GAS5) in SCI-induced neuronal apoptosis remains unclear.
AIM To investigate the role of lncRNA GAS5 in SCI-induced neuronal apoptosis via its interaction with microRNA (miR)-21 and the phosphatase and tensin homolog (PTEN)/AKT pathway.
METHODS SCI rat models and hypoxic neuronal cell models were established. Motor function was assessed using the Basso-Beattie-Bresnahan score. Expression levels of GAS5, miR-21, PTEN, caspase 3, B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), and AKT were measured using quantitative PCR or Western blot analysis. Neuronal apoptosis was determined by TUNEL staining. Dual-luciferase reporter assays validated GAS5-miR-21 binding. Knockdown and overexpression experiments explored the functional effects of the GAS5/miR-21 axis.
RESULTS GAS5 was significantly upregulated in the spinal cord following SCI, coinciding with increased neuronal apoptosis and decreased AKT activation. In vitro experiments demonstrated that GAS5 acted as a molecular sponge for miR-21, leading to increased PTEN expression and inhibition of the AKT signaling pathway, thereby promoting apoptosis. In vivo, GAS5 knockdown attenuated neuronal apoptosis, enhanced AKT activation, and improved motor function recovery in SCI rats.
CONCLUSION GAS5 promotes neuronal apoptosis in SCI by binding to miR-21 and upregulating PTEN expression, inhibiting the AKT pathway. Targeting GAS5 may represent a novel therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Ying-Jie Wang
- Department of Spine Surgery, East Hospital Affiliated to Tongji University School of Medicine, Shanghai 200120, China
| | - Zhong-Zheng Zhi
- Department of Spine Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200434, China
| | - Tao Liu
- Department of Spine Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200434, China
| | - Jian Kang
- Department of Spine Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200434, China
| | - Guang-Hui Xu
- Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| |
Collapse
|
36
|
Chen N, Li Y, Li X. Dynamic role of long noncoding RNA in liver diseases: pathogenesis and diagnostic aspects. Clin Exp Med 2025; 25:160. [PMID: 40369230 PMCID: PMC12078412 DOI: 10.1007/s10238-025-01678-z] [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: 03/10/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Liver disease (LD) is complex pathological condition that has emerged as a major threat to human health and the quality of life. Nonetheless, the molecular mechanisms underlying the pathogenesis of LD have not yet been fully elucidated. Recently, a large amount of evidence has shown that long noncoding RNAs (lncRNAs) play important roles in diverse biological processes in the liver. The dysregulation of lncRNAs in the liver, for example, can affect tumor proliferation, migration, and invasion, contribute to hepatic metabolism disorder of lipid and glucose, and shape of hepatic tumoral microenvironment. Thus, a comprehensive understanding of the functional roles of lncRNAs in LD pathogenesis may provide new perspectives for the development of novel diagnostic and therapeutic tools. In the present review, we summarize the current findings on the relationship between lncRNAs and LD, including the modes of action of lncRNAs, the biological significance of lncRNAs in the pathogenesis of LD, especially in hepatocellular carcinoma (HCC) as well as in some non-neoplastic disorders, and the potential use of lncRNAs as diagnostic biomarkers and therapeutic targets for LD.
Collapse
Affiliation(s)
- Ningning Chen
- Department of Neonatology, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), No. 23976, Jingshi Road, Jinan, 250022, Shandong, China
- School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yunxia Li
- Department of Neonatology, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), No. 23976, Jingshi Road, Jinan, 250022, Shandong, China
| | - Xiaoying Li
- Department of Neonatology, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), No. 23976, Jingshi Road, Jinan, 250022, Shandong, China.
| |
Collapse
|
37
|
Wang B, Liu ZH, Li JJ, Xu JX, Guo YM, Zhang JX, Chu T, Feng ZF, Jiang QY, Wu DD. Role of ferroptosis in breast cancer: Molecular mechanisms and therapeutic interventions. Cell Signal 2025; 134:111869. [PMID: 40379233 DOI: 10.1016/j.cellsig.2025.111869] [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: 03/31/2025] [Revised: 05/04/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Ferroptosis, an iron-dependent cell death pathway distinct from apoptosis, is crucial in breast cancer (BC) research, especially for overcoming resistance in triple-negative breast cancer (TNBC). Unlike traditional apoptosis, ferroptosis involves the glutathione (GSH)/glutathione peroxidase 4 (GPX4) axis, iron-driven oxidative reactions, and phospholipid peroxidation. TNBC, characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), is particularly prone to ferroptosis due to acyl-coenzyme A synthetase (ACSL) 4-related lipid changes and solute carrier family 7 member 11 (SLC7A11)-mediated cystine transport. Recent advancements in biomarkers and therapeutic strategies targeting ferroptosis hold significant promise for the diagnosis and prognosis of TNBC. Notable innovations encompass the development of small-molecule compounds and various methodologies designed to enhance ferroptosis. Combination therapies have demonstrated improved antitumor efficacy by counteracting chemotherapy resistance and inducing immunogenic cell death. Nonetheless, challenges persist in optimizing drug delivery mechanisms and minimizing off-target effects. This review underscores the progress in ferroptosis research and proposes precision oncology strategies that exploit metabolic flexibility in BC, intending to transform TNBC treatment and enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Bo Wang
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Zi-Hui Liu
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Jun-Jie Li
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Jia-Xing Xu
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Ya-Mei Guo
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Jing-Xue Zhang
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Zhi-Fen Feng
- School of Nursing and Health, Henan University, Kaifeng, Henan 475004, China.
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| | - Dong-Dong Wu
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
38
|
Zeng L, Wu Y, Zhu L, He J, Yuan Y, Wang X, Tang K, Tan W. Targeting long non-coding RNA RP11-502I4.3 inhibits the trend of angiogenesis in diabetic retinopathy. PLoS One 2025; 20:e0312791. [PMID: 40367061 PMCID: PMC12077687 DOI: 10.1371/journal.pone.0312791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/25/2025] [Indexed: 05/16/2025] Open
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness. We hypothesised that the long non-coding RNA RP11-502I4.3 may be involved in angiogenesis associated with DR. We investigated the role of RP11-502I4.3 in DR by examining its regulation of vascular endothelial growth factor (VEGF). We assessed differences in RP11-502I4.3 expression between the control group and streptozotocin-induced diabetic rats or high glucose (HG)-stimulated human retinal microvascular endothelial cells (HRMECs). VEGF expression was measured with and without lentiviral vectors overexpressing RP11-502I4.3. We analysed the structural alterations related to DR after overexpressing RP11-502I4.3. Our analysis revealed that RP11-502I4.3 expression was lower in the retinas of diabetic rats and HG-stimulated HRMECs compared with normal glucose conditions. Overexpressing of RP11-502I4.3 resulted in decreased VEGF levels. Diabetic rats exhibited retinopathy characterised by thinning of the retinal layer thickness, structural changes in the inner and outer nuclear layers, a reduced count of retinal ganglion cells, and the presence of acellular capillaries. The proliferative activity, migration count, and tube formation ability of HG-treated HRMECs were significantly higher than those of the control group. However, these changes were inhibited by RP11-502I4.3 overexpression. Overexpression RP11-502I4.3 might inhibit retinopathy of diabetic rats and HG-induced angiogenesis by downregulating VEGF expression.
Collapse
Affiliation(s)
- Lan Zeng
- Department of Ophthalmology, The First People’s Hospital of Zunyi (also known as The Third Affiliated Hospital of Zunyi Medical University), Zunyi, China
| | - Yuhao Wu
- Zunyi Medical University, Zunyi, China
| | | | - Junhao He
- Department of Ophthalmology, The First People’s Hospital of Zunyi (also known as The Third Affiliated Hospital of Zunyi Medical University), Zunyi, China
- Scientific Research Center, The First People’s Hospital of Zunyi (also known as The Third Affiliated Hospital of Zunyi Medical University), Zunyi, China
| | - Yuan Yuan
- Department of Ophthalmology, The First People’s Hospital of Zunyi (also known as The Third Affiliated Hospital of Zunyi Medical University), Zunyi, China
| | - Xiaocong Wang
- Department of Ophthalmology, The First People’s Hospital of Zunyi (also known as The Third Affiliated Hospital of Zunyi Medical University), Zunyi, China
| | - Kai Tang
- Zunyi Medical University, Zunyi, China
- Department of Ophthalmology, Heyou Hospital, Foshan, China.
| | - Wei Tan
- Department of Ophthalmology, The First People’s Hospital of Zunyi (also known as The Third Affiliated Hospital of Zunyi Medical University), Zunyi, China
| |
Collapse
|
39
|
Tang J, Huang Z, Yu P. The role of JPX in regulating FUS/SLC7A11 signaling pathway mediated ferroptosis in keloid fibroblasts and its potential in scar repair. Biochem Biophys Res Commun 2025; 763:151770. [PMID: 40233433 DOI: 10.1016/j.bbrc.2025.151770] [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: 03/26/2025] [Revised: 04/01/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Keloid scar, a fibrotic disease initiated by aberrant fibroblast proliferation, is influenced by ferroptosis. This investigation aims to elucidate the mechanism of lncRNA JPX regulating ferroptosis in keloid fibroblasts. METHODS AND RESULTS We procured 30 samples of keloid tissue and adjacent normal skin tissues from patients undergoing treatment for keloid scars, subsequently isolating fibroblasts from both the keloid lesions and unaffected portions. JPX expression levels in these lesion and keloid fibroblast samples were detected using qRT-PCR. We then validated the regulatory role of JPX on FUS and SLC7A11 through RNA immunoprecipitation and actinomycin D assays. Subsequently, we overexpressed or silenced JPX and/or SLC7A11 in keloid fibroblasts under conditions with or without ferroptosis inhibitor Fer-1, assessing cell viability, migration, invasion, extracellular matrix (ECM) markers via MTT, Transwell and Western blot assays, and evaluating cellular iron content, reactive oxygen species (ROS), malondialdehyde (MDA), and glutathione (GSH) levels as well as the expression of ferroptosis-related proteins to assess ferroptosis. JPX was up-regulated in keloid tissue and keloid fibroblasts. JPX promoted SLC7A11 stability and expression through FUS. JPX silence suppressed viability, migration, invasion and ECM production, yet facilitated ferroptosis of keloid fibroblasts, while these effects could be reversed by Fer-1 or SLC7A11 overexpression. CONCLUSION JPX regulates ferroptosis within fibroblast derived from scar tissue via the FUS/SLC7A11 pathway, demonstrating its potential utility in facilitating scar repair processes.
Collapse
Affiliation(s)
- Jianhui Tang
- Department of Medical Cosmetology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, People's Republic of China.
| | - Zhaoming Huang
- Department of Medical Cosmetology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, People's Republic of China
| | - Panpan Yu
- Department of Medical Cosmetology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, People's Republic of China
| |
Collapse
|
40
|
Najari P, Akbarzadeh S, Rajabi A, Tayefeh-Gholami S, Abbaslou EM, Ghasemzadeh T, Hosseinpourfeizi M, Safaralizadeh R. Evaluation of LncRNAs CBR3-AS1 and PCA3 expression in Gastric cancer and their correlation to clinicopathological variables. Genes Cancer 2025; 16:18-25. [PMID: 40356687 PMCID: PMC12068319 DOI: 10.18632/genesandcancer.241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a multifactorial disease with a high death rate due to the unknown mechanisms involved in the developing, progressing, and late diagnosing GC. Several cancers have been linked to Long non-coding RNAs (lncRNAs), including GC, through differential expression. They play a crucial role in tumorigenesis pathways as modulatory factors, making them intriguing clinical and diagnostic biomarkers for many malignancies. This study's objective is to compare the lncRNAs CBR3-AS1 and PCA3 expression levels in tumoral tissues to marginal tissues and the clinicopathological features of patients. METHODS AND RESULTS 100 GC patients' tumoral and marginal tissue samples from Tabriz's Valiasr Hospital were gathered for this case-control research. To determine the expression level of PCA3 and CBR3-AS1 lncRNAs in GC, total RNA was extracted, and the qRT-PCR technique was employed. Compared to adjacent marginal tissues, the tumor tissue of patients with GC showed a significant increase in the expression levels of PCA3 and CBR3-AS1 (P < 0.0001). The expression ratio of lncRNA CBR3-AS1 and PCA3 did not significantly correlate with clinicopathological variables. The ROC curve's findings lead to the conclusion that the genes lncRNAs PCA3 and CBR3-AS1, with AUC values of 0.68 and 0.79, respectively, suggest that they could play carcinogenic roles in GC and may act as moderate diagnostic biomarkers for GC. CONCLUSIONS In GC, CBR3-AS1 and PCA3 may be utilized as therapeutic targets and prognostic biomarkers, respectively.
Collapse
Affiliation(s)
- Parisa Najari
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Sama Akbarzadeh
- Department of Biophysics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
- Institute of Graduate Studies in Health Sciences, Istanbul University, Istanbul, Turkey
| | - Ali Rajabi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Samaneh Tayefeh-Gholami
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Elaheh Malek Abbaslou
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Tooraj Ghasemzadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
41
|
Liu Y, Li Q, Wu Y, Tang H, Zou C, Dou W, Zhang G. LINC01614 enhances neural invasion and gastric cancer progression by stabilizing VCAM1 and activating the AKT/mTOR pathway. Cell Signal 2025:111860. [PMID: 40349810 DOI: 10.1016/j.cellsig.2025.111860] [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: 02/24/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Gastric cancer (GC) is among the most fatal cancers worldwide. Neural infiltration (NI) is a poorly understood metastatic pathway with limited insights into its regulation and therapeutic targets. METHODS The GEPIA algorithm and RBPsuite analyzed the LINC01614-IGF2BP3-VCAM1 axis in the TCGA-STAD dataset. GC-NI clinical samples and cell lines validated LINC01614 expression and its clinical relevance. CCK-8, colony formation, flow cytometry, Transwell migration and dorsal root ganglia (DRG)-GC co-culture assays assessed LINC01614's role in cell proliferation, apoptosis, migration, invasion and NI. RNA immunoprecipitation, RNA pull-down combined with mass spectrometry, actinomycin D stability assays, and fluorescence in situ hybridization elucidated the formation of an RNA-protein ternary complex and its regulatory effects on VCAM1 stability and the AKT/mTOR pathway. An in vivo murine model was established to examine the impact of LINC01614 knockdown on tumor growth and NI, with tumor progression tracked using IVIS spectral imaging and immunohistochemical staining of GP9.5. RESULTS LINC01614 was significantly overexpressed in GC-NI-positive patients and correlated with poor prognosis, as validated in clinical samples and TCGA-STAD datasets. Functional assays demonstrated that LINC01614 knockdown inhibited GC cell proliferation, migration, invasion, and NI potential. Mechanistically, RNA pull-down followed by mass spectrometry identified IGF2BP3 as a key binding partner of LINC01614. Subsequent RNA immunoprecipitation assays showed that LINC01614 interacts with IGF2BP3 to form an RNA-protein ternary complex with VCAM1 in the nucleus, as further validated by fluorescence in situ hybridization-immunofluorescence. This complex stabilized VCAM1 mRNA, as demonstrated by actinomycin D assays, leading to its upregulation. Enhanced VCAM1 expression activated the AKT/mTOR pathway, which drove tumor progression and NI. Knockdown of LINC01614 disrupted this axis, reduced VCAM1 levels, attenuated AKT/mTOR pathway activation, and mitigated the malignant phenotype of GC cells. CONCLUSION LINC01614 drives NI in GC by stabilizing VCAM1 and activating the AKT/mTOR pathway, making it a promising biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yujing Liu
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215000, China
| | - Qian Li
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215008, China
| | - Yongyou Wu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215008, China
| | - Hongjie Tang
- Department of Surgery, The Fifth People's Hospital of Suzhou / The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215100, China
| | - Chen Zou
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215000, China
| | - Wenhuan Dou
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215000, China
| | - Guoqiang Zhang
- Department of Surgery, The Fifth People's Hospital of Suzhou / The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215100, China.
| |
Collapse
|
42
|
Bai Y, Zhang H, Lin Z, Huang S, Xie F, Gao S, Wong CCL, Wu Y, Wang X, Zhao H, Zhang Y. Lung cancer associated transcript 1 binds heat shock protein 90 to promote growth of hepatocellular carcinoma. Cell Signal 2025; 129:111671. [PMID: 39971224 DOI: 10.1016/j.cellsig.2025.111671] [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: 01/08/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of leading causes of cancer-related death and new approaches are urgently needed, given current dearth of therapies. Long non-coding RNAs (lncRNAs) have been linked to cancer formation and impact cell regulatory pathways. We have investigated molecular mechanisms of action of "lung cancer associated transcript 1" (LUCAT1, a lncRNA) in HCC and studied the potential role of targeting these. We analyzed expression levels of LUCAT1 in TCGA dataset and another 148 HCC patients in Peking Union Medical College Hospital (PUMCH). Expression analysis using TCGA database and patient cohort revealed LUCAT1 to be upregulated in HCC. LUCAT1 levels were closely associated with clinical prognosis. Subcellular localization patterns of LUCAT1 in HCC tissues and cells were identified by RNAscope. Engineered antisense oligonucleotides (ASOs) targeting LUCAT1 were used to test anti-tumor effectiveness using in vitro and in vivo models. CHIRP-MS and RNA pull-down assays were conducted to demonstrate the mechanism of action of LUCAT1.LUCAT1 expression facilitated nuclear accumulation of HSP90. This interaction evoked persistent phosphorylation and constitutive activation of signal transducer and activator of transcription 3 (STAT3), potentially driving HCC growth and metastases. These tumor-promoting effects were substantively diminished using ASOs against LUCAT1, both in vitro and in vivo. LUCAT1 selectively boosts oncogenic property of HSP90 in driving STAT3 activation, which can be effectively and precisely inhibited by designer ASOs. We propose novel potential therapeutic avenues that are selective, cost-effective and seemingly nontoxic.
Collapse
Affiliation(s)
- Yi Bai
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China; Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin 300192, China
| | - Haohai Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China; Division of Gastroenterology, Department of Medicine & Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Zijie Lin
- Tianjin Medical University, Tianjin 300070, China
| | - Shan Huang
- Beijing Institute of Infectious Diseases, Beijing 100015, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease of Ministry of Education, Department of Cell Biology, Tianjin Medical University, Tianjin 300070, China; Institute of Infectious Diseases, National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| | - Fucun Xie
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100021, China
| | - Shuaixin Gao
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), 100730 Beijing, China; Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Catherine C L Wong
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), 100730 Beijing, China; Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, 100084 Beijing, China.
| | - Yan Wu
- Division of Gastroenterology, Department of Medicine & Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Xi Wang
- Beijing Institute of Infectious Diseases, Beijing 100015, China; Institute of Infectious Diseases, National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China.
| | - Yamin Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin 300192, China.
| |
Collapse
|
43
|
Chen X, Li C, Li J, Guo Z, Zhang S, Guo C, Yan H. LncRNA HOTAIR Interaction With WTAP Promotes m6A Methyltransferase Complex Assembly and Posterior Capsule Opacification Formation by Increasing THBS1. Invest Ophthalmol Vis Sci 2025; 66:20. [PMID: 40341312 PMCID: PMC12068528 DOI: 10.1167/iovs.66.5.20] [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: 01/21/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025] Open
Abstract
Purpose To explore the role of long non-coding RNAs (lncRNAs) and N6-methyladenosine (m6A) in posterior capsule opacification (PCO) and their underlying mechanisms. Methods The localization of lncRNAs and proteins was analyzed using fluorescence in situ hybridization and immunofluorescence staining. RNA m6A quantification, RNA immunoprecipitation, co-immunoprecipitation, MeRIP-seq, MeRIP-qPCR, western blotting, wound healing, and Transwell assays were applied to elucidate the underlying mechanisms. Results The levels of lncRNA HOX transcript antisense intergenic RNA (HOTAIR) and m6A methylation increased significantly during epithelial-mesenchymal transition (EMT) in lens epithelial cells (LECs). HOTAIR promoted EMT and m6A methyltransferase activity but had no effect on methyltransferase activity and was not modified by m6A. Nevertheless, HOTAIR interacted with WT1-associated protein (WTAP), a key m6A writer protein, facilitating WTAP-mediated recruitment of METTL3-METTL14 heterodimers and enhancing m6A modification. The HOTAIR/WTAP complex elevated m6A levels, thrombospondin 1 (THBS1) expression, and EMT in LECs. Conclusions LncRNA HOTAIR enhances the assembly of the WTAP/METTL3/METTL14 complex and promotes EMT in LECs by upregulating m6A modification and THBS1 expression. Targeting the HOTAIR/WTAP/THBS1 pathway may prevent or treat PCO.
Collapse
Affiliation(s)
- Xi Chen
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Chenshuang Li
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Jiankui Li
- Department of Gynecology & Obstetrics, The 960th Hospital of PLA, Jinan, Shandong, China
| | - Zaoxia Guo
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Siqi Zhang
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Chenjun Guo
- Department of Ophthalmology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Hong Yan
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| |
Collapse
|
44
|
Xiao Q, Yang S, Yang Y, Ni H, Li Z, Wang C, Liu W, Han Y, Li Y, Zhang Y. LncRNA A2ml2 inhibits fatty liver hemorrhage syndrome progression and function as ceRNA to target LPL by sponging miR-143-5p. Poult Sci 2025; 104:105003. [PMID: 40073631 PMCID: PMC11950757 DOI: 10.1016/j.psj.2025.105003] [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: 12/17/2024] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025] Open
Abstract
Fatty liver hemorrhage syndrome (FLHS) is the most common metabolic diseases in laying hens during the late-laying period, and it causes a significant economic burden on the poultry industry. The competing endogenous RNA plays crucial roles in the occurrence and development of fatty liver. Based on the previously constructed lncRNA-miRNA-mRNA networks, we selected the axis of ENSGALT00000079786-LPL-miR-143-5p for further study to elucidate its mechanistic role in development of fatty liver. In this study, we identified a novel highly conserved lncRNA (ENSGALT00000079786) in poultry, which we designated as lncRNA A2ml2 based on its chromosomal location. Fluorescent in situ hybridization (FISH) revealed that lncRNA A2ml2 was localized in both the nucleus and cytoplasm. Dual-luciferase reporter assay validated the targeted relationship between lncRNA A2ml2, miR-143-5p, and the LPL gene. To further analyze the lncRNA A2ml2 and miR-143-5p function, lncRNA A2ml2 overexpression vector was successfully constructed and transfected into Leghorn male hepatocellular (LMH) cells, which could remarkably inhibit cellular lipid deposition was detected by oil red staining (P < 0.01), the opposite occurred for miR-143-5p (P < 0.01). qPCR demonstrated an inverse correlation between miR-143-5p expression and lncRNA A2ml2 expression, and confirmed that miR-143-5p directly target lncRNA A2ml2. Similarly, we found an inverse correlation between expression of LPL and the expression of miR-143-5p. To further investigate the interactions among these three factors and their effects on cellular lipid metabolism, we assessed the expression levels of LPL by co-transfecting lncRNA A2ml2 with miR-143-5p mimic and miR-143-5p mimic binding site mutants. Co-transfection experiments showed that miR-143-5p diminished the promoting effect of lncRNA A2ml2 on LPL. Meanwhile, miR-143-5p has the capacity to mitigate the suppressive impact of lncRNA A2ml2 overexpression on lipid accumulation in LMH cells. The results revealed that lncRNA A2ml2 attenuated hepatic lipid accumulation through negatively regulating miR-143-5p and enhancing LPL expression in LMH cells. Our findings offer novel insights into ceRNA-mediated in FLHS and identify a novel lncRNA as a potential molecular biomarker.
Collapse
Affiliation(s)
- Qingxing Xiao
- College of Animal Science, Jilin University, Changchun, 130062, China
| | - Sibao Yang
- Department of Cardiovascular Medicine, China-Japan Union Hospital of Jilin University, No.126 Xiantai street of Changchun city, Jilin, 130031, China
| | - Yuwei Yang
- College of Animal Science, Jilin University, Changchun, 130062, China
| | - Hongyu Ni
- College of Animal Science, Jilin University, Changchun, 130062, China
| | - Zongdi Li
- College of Animal Science, Jilin University, Changchun, 130062, China
| | - Chengwen Wang
- College of Animal Science, Jilin University, Changchun, 130062, China
| | - Wuyang Liu
- College of Animal Science, Jilin University, Changchun, 130062, China
| | - Yuxin Han
- College of Animal Science, Jilin University, Changchun, 130062, China
| | - Yumei Li
- College of Animal Science, Jilin University, Changchun, 130062, China
| | - Yonghong Zhang
- College of Animal Science, Jilin University, Changchun, 130062, China.
| |
Collapse
|
45
|
Wang Q, Sun Z, Guo J, Li H, Zhang J, Zhang B, Zhou B, Feng Y. Tumor-derived exosomal LINC01812 induces M2 macrophage polarization to promote perineural invasion in cholangiocarcinoma. Cancer Lett 2025; 617:217596. [PMID: 40081462 DOI: 10.1016/j.canlet.2025.217596] [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: 01/08/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
M2 macrophages play a critical role in the tumor microenvironment of invasive solid tumors. They are closely associated with perineural invasion (PNI) and are often linked to poor prognosis. In this context, tumor-derived exosomes serve as important mediators of intercellular communication. However, the relationship between tumor cell-induced M2 macrophages and PNI in cholangiocarcinoma remains unexplored. In this study, we utilized multiplex immunofluorescence and transcriptomic sequencing to demonstrate the upregulation of LINC01812 in cholangiocarcinoma tissues and its positive correlation with M2 macrophage infiltration. Exosomal lncRNA sequencing, exosome uptake experiments, RNA pull-down assays, and mass spectrometry analysis demonstrated that macrophages can internalize exosomal LINC01812 and promote the M2 phenotype in cholangiocarcinoma cells. Additionally, Transwell and in vitro cocultures with the dorsal root ganglia confirmed that the tumor microenvironment significantly enhances the nerve infiltration of cholangiocarcinoma cells via M2 macrophages. The findings of this study indicate that exosomes containing LINC01812 derived from cholangiocarcinoma can induce M2 macrophage polarization and facilitate nerve infiltration, thereby providing new potential therapeutic targets for managing PNI in cholangiocarcinoma.
Collapse
Affiliation(s)
- Qinlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Zhaowei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Jingyun Guo
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Haoran Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Jingru Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Bingyuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Yujie Feng
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| |
Collapse
|
46
|
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
|
47
|
Shao Y, Gu Q, Yuan Y, Wang L, Yu T. The Preconditioning with Sevoflurane Alleviates Hypoxia-Reoxygenation-Induced Myocardial Cell Injury by Regulating the lncRNA LINC00265/miR-370-3p Axis. Cardiovasc Toxicol 2025; 25:778-789. [PMID: 40111712 DOI: 10.1007/s12012-025-09984-4] [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: 10/11/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
In recent years, the cardioprotective effects of the volatile anesthetic sevoflurane (SEV) have been confirmed, yet its underlying molecular mechanisms remain incompletely elucidated. Notably, lncRNA LINC00265 has been identified as dysregulated in damaged cardiomyocytes, potentially contributing to disease progression. However, limited research has focused on the interplay between SEV and lncRNA LINC00265. The main objective of this study was to explore the mechanism and role of lncRNA LINC00265 in mediating the cardioprotective effects of SEV against myocardial injury. An in vitro hypoxia/reoxygenation (H/R) model was created in AC16 cells following pretreatment with varying concentrations of SEV. RT-qPCR was used to evaluate the levels of lncRNA LINC00265, miR-370-3p, IL-6, and TNF-α. The concentrations of CK-MB and cTnI were determined using ELISA. Cell viability was evaluated using CCK-8, and apoptosis was quantified by flow cytometry. Additionally, the relationship between lncRNA LINC00265 and miR-370-3p was confirmed using a dual-luciferase reporter assay. Prolonged hypoxia gradually rose in lncRNA LINC00265 levels, which was reversed by SEV pretreatment. SEV pretreatment mitigated H/R-induced decreases in cell viability, increases in apoptosis, and excessive production of IL-6, TNF-α, CK-MB, and cTnI. However, the protective effects of SEV were counteracted by lncRNA LINC00265 overexpression. A negative regulatory relationship between lncRNA LINC00265 and miR-370-3p was discovered. miR-370-3p overexpression mitigated diminished protective effects of SEV by elevated lncRNA LINC00265 in myocardial injury. lncRNA LINC00265 could diminish the protective effects of SEV against myocardial injury by functioning as a sponge for miR-370-3p.
Collapse
Affiliation(s)
- Yangge Shao
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Qiang Gu
- Department of Cardiology, Qianjiang Hospital, Renmin Hospital of Wuhan University, The Affiliated Qianjiang Central Hospital of Yangtze University, Qianjiang, 433100, China
| | - Yawei Yuan
- Department of Anesthesiology, Ruijin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Long Wang
- Department of Pain Medicine, The First Medical Center, Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing, 100853, China.
| | - Taowei Yu
- Department of Medical Laboratory, Dianjiang People's Hospital of Chongqing, No. 116, North Street, Guixi Street, Dianjiang County, Chongqing, 408300, China.
| |
Collapse
|
48
|
Xu C, Yu XH, Wang G, Luo W, Chen L, Xia XD. The m 7G methylation modification: An emerging player of cardiovascular diseases. Int J Biol Macromol 2025; 309:142940. [PMID: 40210060 DOI: 10.1016/j.ijbiomac.2025.142940] [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: 11/27/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Cardiovascular diseases severely endanger human health and are closely associated with epigenetic dysregulation. N7-methylguanosine (m7G), one of the common epigenetic modifications, is present in many different types of RNA molecules and has attracted significant attention due to its impact on various physiological and pathological processes. Recent studies have demonstrated that m7G methylation plays an important role in the occurrence and development of multiple cardiovascular diseases. Application of small molecule inhibitors to target m7G modification mediated by methyltransferase-like protein 1 (METTL1) has shown potentiality in the treatment of cardiovascular diseases. In this review, we summarize the basic knowledge about m7G modification and discuss its role and therapeutic potential in diverse cardiovascular diseases, aiming to provide a theoretical foundation for future research and therapeutic intervention.
Collapse
Affiliation(s)
- Can Xu
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, China
| | - Gang Wang
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Wei Luo
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Lei Chen
- Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, China.
| | - Xiao-Dan Xia
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
| |
Collapse
|
49
|
Quan Y, Gao S. LncRNA HOXA-AS2 Can Predict the Risk of Acute Respiratory Distress Syndrome and 28-Day Mortality in Patients With Sepsis. THE CLINICAL RESPIRATORY JOURNAL 2025; 19:e70082. [PMID: 40396530 DOI: 10.1111/crj.70082] [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] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVE This study aimed to explore the diagnostic and predictive value of lncRNA HOXA-AS2 for acute respiratory distress syndrome (ARDS) and 28-day mortality in sepsis patients. METHODS The levels of HOXA-AS2 in sepsis and ARDS patients were detected by real-time quantitative reverse transcription PCR (RT-qPCR). The receiver operating curve (ROC) curve was used to evaluate the diagnostic value of HOXA-AS2 for sepsis and ARDS. The K-M curve was used to evaluate the effect of HOXA-AS2 on the prognosis. Logistic regression analysis and COX regression analysis were used to explore the risk factors influencing ARDS and death. Additionally, an ARDS cell model was constructed to explore the effects of HOXA-AS2 on cell viability, inflammation, and endothelial glycocalyx. RESULTS HOXA-AS2 decreased in sepsis patients who developed ARDS and died. This molecule can not only serve as a diagnostic marker for sepsis but also act as a risk factor to predict the risk of ARDS and death within 28 days in patients with sepsis. Sepsis patients with low levels of HOXA-AS2 are more prone to ARDS and death. In cells attacked by lipopolysaccharide (LPS), overexpression of HOXA-AS2 inhibited apoptosis, inflammation, and the degradation of endothelial glycocalyx. CONCLUSION In sepsis patients, HOXA-AS2 has the potential to serve as a predictive marker for ARDS and 28-day mortality. This molecule may delay the progression of ARDS by inhibiting inflammation and the degradation of the endothelial glycocalyx.
Collapse
Affiliation(s)
- Youhong Quan
- Intensive Care Unit, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, China
| | - Song Gao
- Intensive Care Unit, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, China
| |
Collapse
|
50
|
Kong S, Li J, Pan X, Zhao C, Li Y. ZNF384 and m6A methylation promote the progression of hepatocellular carcinoma by regulating the interaction between LINC00342 and DAPK1. Cell Signal 2025; 129:111666. [PMID: 39961407 DOI: 10.1016/j.cellsig.2025.111666] [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: 09/24/2024] [Revised: 12/05/2024] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality. Many lncRNAs play important regulatory roles in the pathogenesis of HCC, but the mechanism of action of LINC00342 in the progression of HCC remains unclear. In this study, we assessed 24 pairs of HCC tissues and adjacent normal tissues as well as HCC cells and a nude mouse model of HCC. Gene and protein expression was evaluated by flow cytometry, CCK-8, RIP, colony formation assay, and TUNEL staining. This study revealed that LINC00342 was highly expressed in HCC tissues and cells. LINC00342 knockdown significantly inhibited the proliferation and migration of HCC cells, promoted apoptosis, inhibited tumor growth in vivo, and increased the sensitivity of HCC cells to cisplatin. The opposite effect was observed in LINC00342-overexpressing cells. Mechanistically, ZNF384 and m6A methylation can promote the transcription and stability of LINC00342, and LINC00342 can bind to DAPK1, which inhibits Cyt C release and the activation of caspase family proteins to accelerate HCC progression. Our study indicated that the inhibition of LINC00342 expression may represent a new breakthrough for HCC treatment.
Collapse
Affiliation(s)
- Shujia Kong
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Jiaxun Li
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Xin Pan
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Chen Zhao
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Yanwen Li
- Intensive Care Unit, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China.
| |
Collapse
|